1
|
Stoljar A, Zarodniuk M, Bichele R, Armulik EH, Haljasorg U, Humeau R, Besnard M, Haljasmägi L, Tserel L, Peltser M, Salumets A, Kekäläinen E, Kisand K, Guillonneau C, Laan M, Peterson P. Impaired Aire-dependent IFN signaling in the thymus precedes the protective autoantibodies to IFNα. J Exp Med 2025; 222:e20241403. [PMID: 40304722 PMCID: PMC12042843 DOI: 10.1084/jem.20241403] [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: 08/08/2024] [Revised: 01/28/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Recent studies have highlighted the role of the thymus in maintaining immune tolerance to type 1 interferons (T1 IFNs). Individuals with thymic abnormalities, such as autoimmune regulator (AIRE) gene mutations, frequently develop neutralizing autoantibodies to interferon-alpha (IFNα). Unlike mice, Aire-deficient rats develop robust autoantibodies to IFNα. Using this rat model, we show that Aire regulates the thymic expression of interferon-stimulated genes (ISGs), which occurs before developing anti-IFNα autoantibodies. In the periphery, we observed a widespread downregulation of ISGs across immune cells and reduced activation of natural killer (NK) cells. Furthermore, the presence of anti-IFNα autoantibodies correlated with reduced peripheral tissue inflammation, suggesting their role in dampening T1 IFN signaling and minimizing tissue infiltration. Our findings reveal that Aire-mediated regulation of thymic T1 IFN signaling is linked to the production of protective anti-IFNα autoantibodies, which inversely correlate with autoimmune pathology in peripheral tissues.
Collapse
Affiliation(s)
- Artur Stoljar
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Maksym Zarodniuk
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Rudolf Bichele
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Elise Helene Armulik
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Uku Haljasorg
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Romain Humeau
- Nantes University, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, CNRS, Nantes, France
| | - Marine Besnard
- Nantes University, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, CNRS, Nantes, France
| | - Liis Haljasmägi
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Liina Tserel
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Merili Peltser
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Ahto Salumets
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Eliisa Kekäläinen
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kai Kisand
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Carole Guillonneau
- Nantes University, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, CNRS, Nantes, France
| | - Martti Laan
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Pärt Peterson
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
2
|
Zhou J, Gao T, Tang W, Wang Z, Zhao L, Wang L. Cyclophilin D knockdown/knockout promotes microglia M2 polarization by inhibiting STAT1 to alleviate neuroinflammation in neonatal white matter injury. Brain Res 2025; 1856:149596. [PMID: 40132721 DOI: 10.1016/j.brainres.2025.149596] [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: 01/07/2025] [Revised: 03/08/2025] [Accepted: 03/21/2025] [Indexed: 03/27/2025]
Abstract
The activation of microglia cells is intimately associated with the pathophysiology of neuroinflammation and neonatal white matter injury (WMI). Cyclophilin D (CypD), a matrix cyclophilin, is known to be one of the important regulators of mitochondrial permeability transition pore. Currently, CypD has been discovered the function of regulating inflammation. However, its impact on microglia in the context of neonatal WMI remains unclear. In our study, CypD inhibition ameliorated microglia activation, decreased pro-inflammatory factor levels, and increased anti-inflammatory factor levels in both neonatal WMI mice and oxygen glucose deprivation/reperfusion (OGD/R)-induced BV2 microglial cells. CypD knockout promoted myelination and rescued neurological function in mice following hypoxic-ischemic injury. In addition, CypD knockdown alleviated mitochondrial dysfunction of BV2 microglial cells. RNA-Seq indicated that CypD inhibition downregulated STAT1. Western blotting results verified that CypD inhibition significantly downregulated the phosphorylation level of STAT1. Our research revealed the protective role of CypD inhibition in neuroinflammation and mitochondrial function of microglia. Targeting CypD expression in microglia may be a potential therapeutic option for neonatal WMI.
Collapse
Affiliation(s)
- Jiayu Zhou
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Ting Gao
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong Province 510623, China
| | - Wan Tang
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Ziming Wang
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Ling Zhao
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Laishuan Wang
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai 201102, China.
| |
Collapse
|
3
|
Daunov M, Klisovic RB. Pegylated Interferons: Still a Major Player for the Treatment of Myeloproliferative Neoplasms. Am Soc Clin Oncol Educ Book 2025; 45:e473912. [PMID: 40305740 DOI: 10.1200/edbk-25-473912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Over the past 35 years, interferons have been explored in various formulations for the management of Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs), such as essential thrombocythemia (ET), polycythemia vera (PV), and myelofibrosis, and remain a key tool in caring for patients with these diseases. These agents are excellent cytoreductive agents with high rates of hematologic response, are helpful in symptom management, and have a long track record of safety and manageable toxicities. More recently, they have shown promise in sustaining responses over many years, with associated reductions in driver mutations (JAK2, MPL, CALR) of these diseases, particularly in PV and ET. Since reductions in molecular mutant allele burden have been correlated with several response outcomes such as reductions in both thrombotic risk and disease progression, there is emerging proof that interferons may offer disease-modifying activity. These long-term benefits and their use as the preferred agent in young pregnant women who need cytoreduction make interferons often the first choice in young adult population who harbor a lifetime risk of progression. Looking forward, the prospect of sustained treatment-free responses, like chronic myeloid leukemia after deep molecular response, and normal life expectancy may also be on the frontier. Despite relative rookies such as JAK inhibitors in the MPN landscape, the veteran in the game, interferon, remains a key player.
Collapse
Affiliation(s)
- Michael Daunov
- Division of Hematology and Oncology, University Hospitals-Seidman Cancer Center, Cleveland, OH
| | - Rebecca B Klisovic
- Division of Hematology and Oncology, University Hospitals-Seidman Cancer Center, Cleveland, OH
| |
Collapse
|
4
|
Duarte-Silva M, Rao M. A gut-wrenching tale of neuronal distress. Neuron 2025; 113:1291-1293. [PMID: 40339562 DOI: 10.1016/j.neuron.2025.03.033] [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: 03/27/2025] [Revised: 03/29/2025] [Accepted: 03/31/2025] [Indexed: 05/10/2025]
Abstract
Neurons innervating the gut are on the frontlines of host-microbe interactions and thus exposed to a myriad of inflammatory and infectious insults. In this issue of Neuron, Forster, Jakob et al.1 reveal that diverse populations of gut-innervating neurons exhibit conserved responses to inflammation, linking interferon signaling to ferroptosis.
Collapse
Affiliation(s)
- Murillo Duarte-Silva
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | - Meenakshi Rao
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Syed F, Ballew O, Lee CC, Rana J, Krishnan P, Castela A, Weaver SA, Chalasani NS, Thomaidou SF, Demine S, Chang G, Coomans de Brachène A, Alvelos MI, Vazquez EM, Marselli L, Orr K, Felton JL, Liu J, Kaddis JS, Marchetti P, Zaldumbide A, Scheuner D, Eizirik DL, Evans-Molina C. Pharmacological inhibition of tyrosine protein-kinase 2 reduces islet inflammation and delays type 1 diabetes onset in mice. EBioMedicine 2025:105734. [PMID: 40335415 DOI: 10.1016/j.ebiom.2025.105734] [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: 09/17/2024] [Revised: 02/20/2025] [Accepted: 04/14/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Tyrosine protein-kinase 2 (TYK2) mediates inflammatory signalling through multiple cytokines, including interferon-α (IFNα), interleukin (IL)-12, and IL-23. TYK2 missense mutations protect against type 1 diabetes (T1D), and inhibition of TYK2 shows promise in other autoimmune conditions. METHODS We evaluated the effects of specific TYK2 inhibitors (TYK2is) in pre-clinical models of T1D, including human β cells, cadaveric islets, iPSC-derived islets, and mouse models. FINDINGS In vitro studies showed that TYK2is prevented IFNα-induced β cell HLA class I up-regulation, endoplasmic reticulum stress, and chemokine production. In co-culture studies, pre-treatment of β cells with TYK2i prevented IFNα-induced antigenic peptide presentation and alloreactive and autoreactive T cell degranulation. In vivo administration of BMS-986202 in two mouse models of T1D (RIP-LCMV-GP and NOD mice) reduced systemic and tissue-localised inflammation, prevented β cell death, and delayed T1D onset. Transcriptional phenotyping of pancreatic islets, pancreatic lymph nodes, and spleen highlighted a role for TYK2 inhibition in modulating signalling pathways associated with inflammation, translational control, stress signalling, secretory function, immunity, and diabetes. Additionally, TYK2i treatment changed the composition of innate and adaptive immune cell populations in the blood and disease target tissues. INTERPRETATION These findings indicate that TYK2i has beneficial effects on both the immune and endocrine compartments in models of T1D, thus supporting a path forward for testing TYK2is in human T1D. FUNDING This work was supported by the National Institutes of Health (NIH), Veteran Affairs (VA), Breakthrough T1D, and gifts from the Sigma Beta Sorority, the Ball Brothers Foundation, and the George and Frances Ball Foundation.
Collapse
Affiliation(s)
- Farooq Syed
- Indiana University School of Medicine, Indianapolis, IN, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Diabetes-Immunology, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Olivia Ballew
- Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Chih-Chun Lee
- Indiana University School of Medicine, Indianapolis, IN, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jyoti Rana
- Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Preethi Krishnan
- Indiana University School of Medicine, Indianapolis, IN, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Angela Castela
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Staci A Weaver
- Indiana University School of Medicine, Indianapolis, IN, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Sofia F Thomaidou
- Department of Cell and Chemical Biology, Leiden University Medical Center, the Netherlands
| | - Stephane Demine
- Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Garrick Chang
- Department of Physics, Indiana University Indianapolis, Indianapolis, IN, USA
| | | | - Maria Ines Alvelos
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Eugenia Martin Vazquez
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Lorella Marselli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Kara Orr
- Indiana University School of Medicine, Indianapolis, IN, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jamie L Felton
- Indiana University School of Medicine, Indianapolis, IN, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jing Liu
- Department of Physics and Astronomy, Purdue University, West Lafayette, IN, USA
| | - John S Kaddis
- Department of Diabetes and Cancer Discovery Science, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Arnaud Zaldumbide
- Department of Cell and Chemical Biology, Leiden University Medical Center, the Netherlands
| | | | - Decio L Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium.
| | - Carmella Evans-Molina
- Indiana University School of Medicine, Indianapolis, IN, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA; Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA.
| |
Collapse
|
6
|
Chen T, Li X, Hou P, He H, Wang H. VAPA suppresses BEFV and VSV-induced type I IFNs signaling response by targeting JAK1 for NEDD4-mediated ubiquitin-proteasome degradation. Vet Microbiol 2025; 304:110456. [PMID: 40080976 DOI: 10.1016/j.vetmic.2025.110456] [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: 12/14/2024] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
VAMP-associated protein A (VAPA) binds to various proteins involved in multiple cellular processes, however, its role in the regulation of type I interferons (IFN-I) signaling has not been elucidated. In this study, we demonstrate that VAPA negatively regulates the IFN-I signaling during bovine epidemic fever virus (BEFV) and vesicular stomatitis virus (VSV) infection. Upon treatment with IFN-β, VAPA negatively regulates the JAK-STAT signaling pathway. Further studies show that VAPA inhibits the IFN-I signaling by promoting the degradation of JAK1 through the ubiquitin-proteasome system during BEFV and VSV infection. Mechanistically, VAPA facilitates the interaction between the E3 ubiquitin ligase NEDD4 and JAK1, thereby enhancing the ubiquitination and subsequent degradation of JAK1. Furthermore, viral titers are markedly reduced, and the promoting effect of VAPA on VSV or BEFV replication is attenuated in NEDD4-deficient cells. Taken together, our findings reveal a novel role for VAPA in negatively regulating the IFN-I signaling response and provide a molecular basis for the design of targeted antiviral agents.
Collapse
Affiliation(s)
- Tianhua Chen
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Xingyu Li
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Peili Hou
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan 250358, China.
| | - Hongbin He
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan 250358, China.
| | - Hongmei Wang
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan 250358, China.
| |
Collapse
|
7
|
Feng C, Li Q, Miao D, Hu X, Huang J, Peng D, Song Y, Zhang D. Mouse models of Tembusu virus infection for differentiating between cluster 2.1 and 2.2 isolates. Vet Microbiol 2025; 304:110474. [PMID: 40101376 DOI: 10.1016/j.vetmic.2025.110474] [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: 11/01/2024] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/20/2025]
Abstract
Tembusu virus (TMUV) cluster 2.1 and 2.2 strains are known to produce lethal neurological disease in mice inoculated by intracerebral (ic) route. Here, we report the comparative clinicopathological findings following experimental infections of 3-week-old BALB/c and Kunming mice with cluster 2.1 isolate H and cluster 2.2 isolate Y. When infected by the subcutaneous (sc) route, both isolates failed to induce disease in mice. When infected by the ic route, both isolates caused lethal neurological disease in mice, with isolate H presenting markedly higher neurovirulence than isolate Y. Further studies with the Kunming mouse model showed that following sc inoculation, both H and Y isolates failed to replicate in brain and spleen, and that following ic inoculation, isolate H replicated to higher levels in brain and spleen than isolate Y. The findings may help to explain non-neuroinvasive property of clusters 2.1 and 2.2 and suggest that enhanced neurovirulence of cluster 2.1 relative to cluster 2.2 is associated with more efficient replication in the central nervous system and in the periphery. Moreover, isolate H induced significantly higher levels of IFN-β, IL-1β, IL-6, TNF-α, Ifit1, and Ifit2 expression relative to isolate Y, indicating a positive correlation between TMUV neurovirulence and magnitude of antiviral innate immune response. The present work demonstrates that the mouse models allow to differentiate between cluster 2.1 and 2.2 isolates and provides mechanistic insights into TMUV-induced disease.
Collapse
Affiliation(s)
- Chonglun Feng
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China.
| | - Qiong Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China.
| | - Dongying Miao
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China.
| | - Xiaoyang Hu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China.
| | - Jingjing Huang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China.
| | - Duo Peng
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China.
| | - Yinuo Song
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China.
| | - Dabing Zhang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China.
| |
Collapse
|
8
|
Li R, Zheng W, Xiao Y, Yu X, Sheng J, Zhang H, Chen C, Ma Z, Wang Y. Mycoplasma hyopneumoniae nuclease Mhp597 negatively regulates TBK1-IRF3-IFN-I pathway by targeting vimentin to facilitate infection. Int J Biol Macromol 2025; 306:141351. [PMID: 39988178 DOI: 10.1016/j.ijbiomac.2025.141351] [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: 11/13/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Infection with Mycoplasma hyopneumoniae (M. hyopneumoniae) leads to chronic infectious pneumonia in pigs, resulting in significant distress and economic losses in the global pig industry. The pathogen secretes various proteins, including toxins, adhesins, and virulence-related enzymes, which facilitate adhesion, invasion, and immune evasion processes between bacteria and the host. However, the effector proteins of M. hyopneumoniae are predominantly uncharacterized. In this study, we demonstrate that the nuclease Mhp597 functions as a potential effector protein of M. hyopneumoniae, and we elucidate its mechanism of action in facilitating immune evasion. Our findings indicate that Mhp597 exhibits high expression efficiency in host cells and significantly inhibits IFN-α and IFN-β protein expression. Using yeast two-hybrid and co-immunoprecipitation experiments, we established that Mhp597 interacts with porcine alveolar macrophage vimentin (Vim) via specific amino acid residues (Arg 232, Lys 256, Phe 263, and Lys 317). Further analysis revealed that Mhp597 inhibited the phosphorylation of TBK1 and IRF3 via Vim, thereby suppressing type I interferon (IFN-I) production and promoting the proliferation of M. hyopneumoniae within host cells. In conclusion, this study provides the first detailed account of the molecular mechanism by which Mhp597 negatively regulates the TBK1-IRF3-IFN-I signaling pathway through Vim, thus facilitating immune evasion and proliferation of M. hyopneumoniae within host cells. These findings enhance our understanding of the pathogenic mechanisms of M. hyopneumoniae and suggest potential molecular targets for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Ruirui Li
- College of Animal Science and Technology, Shihezi University, Shihezi, China; Collaborative Innovation Center for Sheep Health Breeding and Zoonotic Disease Prevention and Control, Shihezi, Xinjiang, China; International Joint Research Center for Animal Health, Shihezi, Xinjiang, China
| | - Wei Zheng
- College of Animal Science and Technology, Shihezi University, Shihezi, China; Collaborative Innovation Center for Sheep Health Breeding and Zoonotic Disease Prevention and Control, Shihezi, Xinjiang, China; International Joint Research Center for Animal Health, Shihezi, Xinjiang, China
| | - Yangyang Xiao
- College of Animal Science and Technology, Shihezi University, Shihezi, China; Collaborative Innovation Center for Sheep Health Breeding and Zoonotic Disease Prevention and Control, Shihezi, Xinjiang, China; International Joint Research Center for Animal Health, Shihezi, Xinjiang, China
| | - Xiaojiao Yu
- College of Animal Science and Technology, Shihezi University, Shihezi, China; Collaborative Innovation Center for Sheep Health Breeding and Zoonotic Disease Prevention and Control, Shihezi, Xinjiang, China; International Joint Research Center for Animal Health, Shihezi, Xinjiang, China
| | - Jinliang Sheng
- College of Animal Science and Technology, Shihezi University, Shihezi, China; Collaborative Innovation Center for Sheep Health Breeding and Zoonotic Disease Prevention and Control, Shihezi, Xinjiang, China; International Joint Research Center for Animal Health, Shihezi, Xinjiang, China
| | - Hui Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi, China; Collaborative Innovation Center for Sheep Health Breeding and Zoonotic Disease Prevention and Control, Shihezi, Xinjiang, China; International Joint Research Center for Animal Health, Shihezi, Xinjiang, China
| | - Chuangfu Chen
- College of Animal Science and Technology, Shihezi University, Shihezi, China; Collaborative Innovation Center for Sheep Health Breeding and Zoonotic Disease Prevention and Control, Shihezi, Xinjiang, China; International Joint Research Center for Animal Health, Shihezi, Xinjiang, China.
| | - Zhongchen Ma
- College of Animal Science and Technology, Shihezi University, Shihezi, China; Collaborative Innovation Center for Sheep Health Breeding and Zoonotic Disease Prevention and Control, Shihezi, Xinjiang, China; International Joint Research Center for Animal Health, Shihezi, Xinjiang, China.
| | - Yong Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, China; Collaborative Innovation Center for Sheep Health Breeding and Zoonotic Disease Prevention and Control, Shihezi, Xinjiang, China; International Joint Research Center for Animal Health, Shihezi, Xinjiang, China.
| |
Collapse
|
9
|
Zhang X, Xiong W, Ma S, Fan S, Sun M, Zhou Q, Yang J, Li J, Zhang X. A novel homozygous ISG15 missense variant leads to severe inflammatory skin lesions, interstitial pneumonia, and basal ganglia calcifications in a Chinese infant with ISG15 deficiency. Gene 2025; 960:149537. [PMID: 40318816 DOI: 10.1016/j.gene.2025.149537] [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: 03/09/2025] [Revised: 04/18/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Type I interferonopathies are a group of rare inherited autoinflammatory disorders characterized by dysregulation of type I interferon (IFN-I) signaling pathways. ISG15, a unique ubiquitin-like (Ubl) modifier in the interferon-stimulated genes (ISGs) family, plays a critical role in innate immune responses induced by IFN-I. When ISG15 function is impaired, it results in a disorder known as ISG15 deficiency, which is classified as an autosomal recessive systemic type I interferonopathy. Here, we report a 4-month-old Chinese patient presenting with inflammatory skin lesions, interstitial pneumonia, and basal ganglia calcifications. Whole-exome sequencing (WES) identified a novel homozygous missense variant (NM_005101.4: exon2: c.392 T > C, p.Leu131Pro) of ISG15. Functional analysis revealed that this variant impaired ISGylation and disrupted the stabilization of USP18, leading to defective negative regulation of IFN-I signaling and consequent excessive IFN-I production. Consistent with this, the patient exhibited elevated expression of ISGs in both peripheral blood and peripheral blood mononuclear cells (PBMCs). Treatment with the Janus kinase (JAK) inhibitor baricitinib rapidly resolved the patient's clinical symptoms. In conclusion, our findings expand the pathogenic spectrum of ISG15 deficiency and highlight the therapeutic efficacy of baricitinib in this disease. Notably, this case represents the first reported instance of a homozygous ISG15 missense variant in the Chinese population and the third such variant reported worldwide, further enriching our understanding of this rare autoinflammatory disease.
Collapse
Affiliation(s)
- Xinyue Zhang
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory for Complex Severe and Rare Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Wen Xiong
- Shenzhen Children's Affiliated Hospital, China Medical University, Shenzhen 518038, China
| | - Shuangyue Ma
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Shiqi Fan
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory for Complex Severe and Rare Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Miao Sun
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory for Complex Severe and Rare Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Qing Zhou
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Jun Yang
- Department of Rheumatology and Immunology, Shenzhen Children's Hospital, Shenzhen 518038, China.
| | - Jianguo Li
- Department of Rheumatology and Immunology, Children's Hospital Affiliated to Capital Institute of Pediatrics, Beijing 100020, China.
| | - Xue Zhang
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory for Complex Severe and Rare Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
10
|
Rao Y, Qin C, Espinosa B, Wang TY, Feng S, Savas AC, Henley J, Comai L, Zhang C, Feng P. Targeting CTP synthetase 1 to restore interferon induction and impede nucleotide synthesis in SARS-CoV-2 infection. mBio 2025:e0064925. [PMID: 40298378 DOI: 10.1128/mbio.00649-25] [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: 02/24/2025] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
Despite the global impact caused by the most recent SARS-CoV-2 pandemic, our knowledge of the molecular underpinnings of its highly infectious nature remains incomplete. We report here that SARS-CoV-2 exploits cellular CTP synthetase 1 (CTPS1) to promote CTP synthesis and suppress interferon (IFN) induction. In addition to catalyzing CTP synthesis, CTPS1 also deamidates interferon regulatory factor 3 (IRF3) to dampen interferon induction. Screening a SARS-CoV-2 expression library, we identified several viral proteins that interact with CTPS1. Functional analyses demonstrate that ORF8 and Nsp8 activate CTPS1 to deamidate IRF3 and negate IFN induction, whereas ORF7b and ORF8 activate CTPS1 to promote CTP synthesis. These results highlight CTPS1 as a signaling node that integrates cellular metabolism and innate immune response. Indeed, small-molecule inhibitors of CTPS1 deplete CTP and boost IFN induction in SARS-CoV-2-infected cells, thus effectively impeding SARS-CoV-2 replication and pathogenesis in mouse models. Our work uncovers an intricate mechanism by which a viral pathogen couples immune evasion to metabolic activation to fuel viral replication. Inhibition of the cellular CTPS1 offers an attractive means to develop antiviral therapy against highly mutagenic viruses.IMPORTANCEOur understanding of the underpinnings of highly infectious SARS-CoV-2 is rudimentary at best. We report here that SARS-CoV-2 activates CTPS1 to promote CTP synthesis and suppress IFN induction, thus coupling immune evasion to activated nucleotide synthesis. Inhibition of the key metabolic enzyme not only depletes the nucleotide pool but also boosts host antiviral defense, thereby impeding SARS-CoV-2 replication. Targeting cellular enzymes presents a strategy to counter the rapidly evolving SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Youliang Rao
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Chao Qin
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Bianca Espinosa
- Department of Chemistry, Dornsife College of Arts, Letters and Sciences, University of Southern California, Los Angeles, California, USA
| | - Ting-Yu Wang
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Shu Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Ali Can Savas
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Jill Henley
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Lucio Comai
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Chao Zhang
- Department of Chemistry, Dornsife College of Arts, Letters and Sciences, University of Southern California, Los Angeles, California, USA
| | - Pinghui Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
11
|
Zhang L, Li B, Liu J, Bian YF, Lin GX, Zhou Y. Unveiling hub genes and biological pathways: A bioinformatics analysis of Trauma-Induced Coagulopathy (TIC). PLoS One 2025; 20:e0322043. [PMID: 40300035 PMCID: PMC12040245 DOI: 10.1371/journal.pone.0322043] [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: 08/23/2024] [Accepted: 03/15/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Trauma-Induced Coagulopathy is a severe condition that rapidly manifests following traumatic injury and is characterized by shock, hypoperfusion, and vascular damage. This study employed bioinformatics methods to identify crucial hub genes and pathways associated with TIC. METHODS Microarray datasets (accession number GSE223245) were obtained from the Gene Expression Omnibus (GEO) database. The data were subjected analyses to identify the Differentially Expressed Genes (DEGs), which were further subjected to GO and KEGG pathway analyses. Subsequently, a Protein-Protein Interaction (PPI) network was constructed and hub DEGs closely linked to TIC were identified using CytoHubba, MCODE, and CTD scores. The diagnostic value of these hub genes was evaluated using Receiver Operating Characteristic (ROC) analysis. RESULTS Among the analyzed genes, 269 were identified as DEGs, comprising 103 upregulated and 739 downregulated genes. Notably, several significant hub genes were associated with the development of TIC, as revealed by bioinformatic analyses. CONCLUSIONS This study highlights the critical impact of newly discovered genes on the development and progression of TIC. Further validation through experimental research and clinical trials is required to confirm these findings.
Collapse
Affiliation(s)
- Lingang Zhang
- Emergency Department, Yuncheng Central Hospital affiliated to Shanxi Medical University,Yuncheng, Shanxi, China
| | - Bo Li
- Reproductive Medicine Department, Yuncheng Central Hospital affiliated to Shanxi Medical University, Yuncheng, Shanxi, China
| | - Jing Liu
- Pathology Department, Yuncheng Central Hospital affiliated to Shanxi Medical University,Yuncheng, Shanxi, China
| | - Yan feng Bian
- Emergency sungery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital,Third Hospital of Shanxi Medical University, China
| | - Guo xing Lin
- Emergency Department, Hebei province Xingtai Third People’s Hospital, Xingtai, China
| | - Ying Zhou
- Emergency Department, Yuncheng Central Hospital affiliated to Shanxi Medical University,Yuncheng, Shanxi, China
| |
Collapse
|
12
|
Ogger PP, Murray PJ. Dissecting inflammation in the immunemetabolomic era. Cell Mol Life Sci 2025; 82:182. [PMID: 40293552 PMCID: PMC12037969 DOI: 10.1007/s00018-025-05715-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/30/2025]
Abstract
The role of immune metabolism, specific metabolites and cell-intrinsic and -extrinsic metabolic states across the time course of an inflammatory response are emerging knowledge. Targeted and untargeted metabolomic analysis is essential to understand how immune cells adapt their metabolic program throughout an immune response. In addition, metabolomic analysis can aid to identify pathophysiological patterns in inflammatory disease. Here, we discuss new metabolomic findings within the transition from inflammation to resolution, focusing on three key programs of immunity: Efferocytosis, IL-10 signaling and trained immunity. Particularly the tryptophan-derived metabolite kynurenine was identified as essential for efferocytosis and inflammation resolution as well as a potential biomarker in diverse inflammatory conditions. In summary, metabolomic analysis and integration with transcriptomic and proteomic data, high resolution imaging and spatial information is key to unravel metabolic drivers and dependencies during inflammation and progression to tissue-repair.
Collapse
Affiliation(s)
- Patricia P Ogger
- Immunoregulation Research Group, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany
| | - Peter J Murray
- Immunoregulation Research Group, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany.
| |
Collapse
|
13
|
Qiao Y, Wei L, Su Y, Tan Q, Yang X, Li S. Nanoparticle-Based Strategies to Enhance the Efficacy of STING Activators in Cancer Immunotherapy. Int J Nanomedicine 2025; 20:5429-5456. [PMID: 40308645 PMCID: PMC12042967 DOI: 10.2147/ijn.s515893] [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: 01/22/2025] [Accepted: 04/16/2025] [Indexed: 05/02/2025] Open
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway plays a critical role in triggering innate and adaptive immune responses through type I interferon activation and immune cell recruitment, holding significant promise for cancer therapy. While STING activators targeting this pathway have been developed, their clinical application is hindered by challenges such as poor membrane permeability, rapid degradation, suboptimal pharmacokinetics, off-target biodistribution, and toxicity. Nanoparticle-based delivery systems offer a promising solution by enhancing the stability, circulation time, tumor accumulation, and intracellular release of STING activators. Furthermore, combining nanoparticle-delivered STING activators with radiotherapy, chemotherapy, phototherapy, and other immunotherapies enables synergistic antitumor effects through multimodal mechanisms, addressing resistance to monotherapies and reducing risks of recurrence and metastasis. This review outlines the immunomodulatory mechanisms of the cGAS-STING pathway, surveys current STING-targeted activators, and comprehensively discusses recent advances in nanoparticle-mediated delivery strategies for STING activation. Additionally, we explore combinatorial approaches that integrate STING-targeted nanotherapies with conventional and emerging treatments. Finally, we highlight the current status, prospects, and challenges of nanoparticle-based STING activation for cancer immunotherapy.
Collapse
Affiliation(s)
- Yi Qiao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Lingyu Wei
- Department of Gynecologic Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, People’s Republic of China
| | - Yinjie Su
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Qinyuan Tan
- Department of Urology, The People’s Hospital of Jimo, Qingdao, People’s Republic of China
| | - Xuecheng Yang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Shengxian Li
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| |
Collapse
|
14
|
Jiang Q, Liu X, Yuan J, Zhang S, Zhang Y, Kan Z, Niu Z, Zhang L, Hu X, Zhou Y, Wang J, Li F, Cao L, Zhang X, Lei C, Song Z. Curcumin activates the JAK-STAT signaling pathway to enhance the innate immune response against porcine epidemic diarrhea virus infection in vivo and in vitro. Vet Microbiol 2025; 305:110535. [PMID: 40286548 DOI: 10.1016/j.vetmic.2025.110535] [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: 02/28/2025] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
Porcine epidemic diarrhea (PED) is a highly pathogenic infectious disease caused by porcine epidemic diarrhea virus (PEDV), which has caused significant economic losses to the global pig industry. Due to the high mutability of the PEDV genome, the development of effective vaccines or drugs to prevent and control PEDV is still facing great difficulties. In this study, we found that the natural compound curcumin showed effective antiviral activity against PEDV in VERO-E6 and IPEC-J2 cells. The Janus Kinase-Signal Transducer and Activator of Transcription (JAK-STAT) signaling pathway was screened by transcriptomics as a potential innate immune mechanism of IPEC-J2 cells against PEDV infection. For PEDV, a highly pathogenic virus, cellular autoimmune response is not sufficient to fight against its infection. Our results demonstrated that curcumin could exert antiviral effects by enhancing the JAK-STAT cascade reaction mediated by type I interferon IFN-α and IFN-β in IPEC-J2 cells. In vivo experiments further confirmed the protective effect of curcumin on PEDV-infected piglets and its positive regulation of the JAK-STAT signaling pathway. In vivo, curcumin prophylaxis significantly enhanced IFN-α and IFN-β-induced JAK-STAT signaling and the production of interferon-stimulated genes (ISGs), and increased the innate immune response, thus exerting antiviral effects effectively. In conclusion, our data indicate that curcumin can effectively resist PEDV infection in IPEC-J2 cells and piglets, which provides a new reference for the development of anti-PEDV drugs with important application prospects.
Collapse
Affiliation(s)
- Qiancheng Jiang
- College of Veterinary Medicine, Southwest University, Chongqing, China.
| | - Xiangyang Liu
- College of Veterinary Medicine, Southwest University, Chongqing, China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China.
| | - Jianbo Yuan
- College of Veterinary Medicine, Southwest University, Chongqing, China.
| | - Shujuan Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China.
| | - Yiling Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China; College of Animal Science and Technology, Chongqing Three Gorges Vocational College, Chongqing, China.
| | - Zifei Kan
- College of Veterinary Medicine, Southwest University, Chongqing, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Zheng Niu
- College of Veterinary Medicine, Southwest University, Chongqing, China; College of Veterinary Medicine, Northwest A and F University, Shanxi, China.
| | - Li Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China.
| | - Xia Hu
- College of Veterinary Medicine, Southwest University, Chongqing, China.
| | - Yang Zhou
- College of Veterinary Medicine, Southwest University, Chongqing, China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China.
| | - Jing Wang
- College of Veterinary Medicine, Southwest University, Chongqing, China.
| | - Fei Li
- College of Veterinary Medicine, Southwest University, Chongqing, China.
| | - Lijing Cao
- Rongchang Vocational Education Center, Chongqing, China.
| | - Xingcui Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China; Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China.
| | - Chenghong Lei
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China.
| | - Zhenhui Song
- College of Veterinary Medicine, Southwest University, Chongqing, China; Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China.
| |
Collapse
|
15
|
Qiu X, Wen R, Wu F, Mao J, Azad T, Wang Y, Zhu J, Zhou X, Xie H, Hong K, Li B, Zhang L, Wen C. The role of double-negative B cells in the pathogenesis of systemic lupus erythematosus. Autoimmun Rev 2025; 24:103821. [PMID: 40274006 DOI: 10.1016/j.autrev.2025.103821] [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: 02/10/2025] [Revised: 04/06/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
B cells are essential to the pathophysiology of systemic lupus erythematosus (SLE), a chronic autoimmune illness. IgD-CD27-double negative B cells (DNB cells) are one of the aberrant B cell subsets linked to SLE that have attracted much scientific interest. There is growing evidence that DNB cells play a significant role in the development of the disease and are strongly linked to the activity of lupus. These cells play a pivotal role in the pathogenesis of SLE by producing a diverse array of autoantibodies, which form immune complexes that drive target organ damage. A comprehensive understanding of SLE pathophysiology necessitates in-depth investigation into DNB cells, not only to elucidate their mechanistic contributions but also to uncover novel therapeutic strategies. According to available data, treatments that target B cells have proven effective in managing SLE; nevertheless, a significant breakthrough in precision medicine for SLE may come from targeting DNB cells specifically. Despite growing interest in DNB cells, their precise characteristics, developmental trajectories, and regulatory mechanisms remain incompletely defined, posing significant challenges to the field. A comprehensive investigation of the regulatory mechanisms governing DNB cell differentiation and expansion in SLE may facilitate novel therapeutic discoveries. This review aims to provide an updated synthesis of current research on DNB cells, with a focus on their origins, developmental trajectories in SLE, and potential as precision therapeutic targets.
Collapse
Affiliation(s)
- Xinying Qiu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China; The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University, Changsha 410007, Hunan, China
| | - RuiFan Wen
- Medical School, Hunan University of Chinese Medicine, No.300 Xueshi Road, Hanpu Science & Education District, Changsha, Hunan 410208, China
| | - Feifeng Wu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Jueyi Mao
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Tasnim Azad
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Yang Wang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Junquan Zhu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Xin Zhou
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Haotian Xie
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Kimsor Hong
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Binbin Li
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Liang Zhang
- The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University, Changsha 410007, Hunan, China; Department of Nephrology, Rheumatology and Immunology, Hunan Children's Hospital, Changsha 410007, Hunan, China.
| | - Chuan Wen
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China.
| |
Collapse
|
16
|
Liu X, Zhang H, Guan S, Tan J, Yeung KWK, Ouyang L, Liu X. Electron Pump and Photon Trap Effect-Derived Selective Antitumor of Fe-Ppy@CaO 2-Modified Polyetheretherketone for Bone Tumor Therapy. ACS NANO 2025; 19:14954-14971. [PMID: 40197016 DOI: 10.1021/acsnano.5c00721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Bone tumors with high mortality and disability have become a major clinical challenge. Herewith, it is necessary to design materials for bone tumor therapy and bone repair. In this work, Fe-doped polypyrrole (Fe-Ppy) and CaO2 are constructed on sulfonated polyetheretherketone (SP) to form a multistage-responsive coating. The coating achieves long-lasting antitumor through chemodynamic therapy (CDT), photothermal therapy (PTT), and combined immunotherapy. Fe-Ppy acts as an electron pump to replenish Fe2+ through oxidizing -NH- to -N+-, which lasts the Fenton reaction and persistently produces reactive oxygen species (ROS) in the tumor microenvironment (TME). CaO2 selectively provides exogenous H2O2 in response to TME to boost the electron cycle. Stronger near-infrared light absorption due to Fe doping and more photon traps caused by porous structure-induced scattering and refraction diminishment improve the photothermal conversion of modified SP. Furthermore, long-lasting ROS and effective photothermal conversion enhance M1 activation to secrete TNF-α and IFN to kill tumor cells. After tumor therapy, Fe-Ppy@CaO2-modified SP could adaptively switch the macrophage to M2 and promote osteogenesis with the abolishment of TME and NIR stimulation. In summary, Fe-Ppy@CaO2-modified SP with long-lasting ROS, enhanced photothermal conversion, and immunomodulation is a potential candidate for bone tumor therapy and tissue repair.
Collapse
Affiliation(s)
- Xingdan Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- Shanghai Key Laboratory of Flexible Medical Robotics, Tongren Hospital, Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200336, China
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Haifeng Zhang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Sub-lane Xiangshan, Hangzhou 310024, China
| | - Shiwei Guan
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Ji Tan
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Kelvin W K Yeung
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Guangdong Engineering Technology Research Center for Orthopaedic Trauma Repair, Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Liping Ouyang
- Shanghai Key Laboratory of Flexible Medical Robotics, Tongren Hospital, Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200336, China
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Sub-lane Xiangshan, Hangzhou 310024, China
| |
Collapse
|
17
|
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
|
18
|
Du Q, He W, Chen X, Liu J, Guan M, Chen Y, Chen M, Yuan Y, Zuo Y, Miao Y, Wang Q, Zhou H, Liu Y, Jiang J, Zheng H. Bilirubin metabolism in the liver orchestrates antiviral innate immunity in the body. Cell Rep 2025; 44:115481. [PMID: 40153433 DOI: 10.1016/j.celrep.2025.115481] [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: 10/06/2024] [Revised: 02/09/2025] [Accepted: 03/07/2025] [Indexed: 03/30/2025] Open
Abstract
Bilirubin metabolism crucially maintains normal liver function, but whether it contributes to antiviral immunity remains unknown. Here, we reveal that the liver bilirubin metabolic pathway facilitates antiviral innate immunity of the body. We discovered that viral infection upregulates uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) expression in the liver, which in turn stabilizes IRF3 proteins to promote type I interferon (IFN-I) production. Moreover, we found that serum unconjugated bilirubin (UCB), a unique physiological substrate of UGT1A1, can competitively inhibit the binding of IFN-I to IFN-I receptor 2 (IFNAR2), thus attenuating IFN-I-induced antiviral signaling of the body. Accordingly, effective bilirubin metabolism in the liver promotes antiviral immunity of the body by specifically employing liver UGT1A1-mediated enhancement of IFN-I production and reducing serum bilirubin-mediated inhibition of IFN-I signaling. This study uncovers the significance of bilirubin metabolism in antiviral innate immunity and demonstrates that conventional IFN-I therapy is less efficient for patients with hepatitis B virus (HBV) with high levels of bilirubin.
Collapse
Affiliation(s)
- Qian Du
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences (IBMS), Collaborative Innovation Center of Hematology, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China
| | - Wei He
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences (IBMS), Collaborative Innovation Center of Hematology, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xiangjie Chen
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences (IBMS), Collaborative Innovation Center of Hematology, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jin Liu
- Department of Infectious Diseases, The Affiliated Infectious Diseases Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Mingcheng Guan
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, China
| | - Yichang Chen
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Meixia Chen
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yukang Yuan
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences (IBMS), Collaborative Innovation Center of Hematology, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China; Department of Laboratory Medicine, Institute of Laboratory Medicine, Translational Clinical Immunology Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, China
| | - Yibo Zuo
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences (IBMS), Collaborative Innovation Center of Hematology, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China; Department of Laboratory Medicine, Institute of Laboratory Medicine, Translational Clinical Immunology Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, China
| | - Ying Miao
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences (IBMS), Collaborative Innovation Center of Hematology, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China; Department of Laboratory Medicine, Institute of Laboratory Medicine, Translational Clinical Immunology Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, China
| | - Qin Wang
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences (IBMS), Collaborative Innovation Center of Hematology, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China; Department of Laboratory Medicine, Institute of Laboratory Medicine, Translational Clinical Immunology Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, China
| | - Haiyan Zhou
- Department of Laboratory Medicine, Institute of Laboratory Medicine, Translational Clinical Immunology Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, China
| | - Yanli Liu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Jingting Jiang
- Department of Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| | - Hui Zheng
- The First Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences (IBMS), Collaborative Innovation Center of Hematology, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu 215123, China; Department of Laboratory Medicine, Institute of Laboratory Medicine, Translational Clinical Immunology Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, China.
| |
Collapse
|
19
|
Marrocco R, Lucero-Meza E, Benedict CA. Type I interferon regulation of group I ILC subsets during both homeostasis and cytomegalovirus infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf051. [PMID: 40258300 DOI: 10.1093/jimmun/vkaf051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/04/2025] [Indexed: 04/23/2025]
Abstract
Type 1 innate lymphoid cells (ILC1s) and conventional natural killer cells belong to the group 1 ILCs (gILC1), characterized largely by T-bet expression and interferon γ secretion. While much has been done to define factors that regulate the development, differentiation, and effector functions of both cell types, little is known about what controls gILC1 homeostasis. Here, mixed bone marrow chimeras were used to define the role of type I interferon receptor (IFNAR) signaling in regulating gILC1 in the spleen and liver at homeostasis and during murine cytomegalovirus infection. We show that basal IFNAR signaling induces cell and tissue-specific phenotypic changes in gILC1, inhibiting bona-fide ILC1 markers (CD49a, CD200R, CXCR6) and regulating expression of perforin and granzymes B and C. Finally, while IFNAR signaling enhances cytokine responsiveness in vitro in both gILC1 subsets, it has a dichotomous effect on interferon γ production during murine cytomegalovirus infection, stimulating it in conventional natural killer cells and inhibiting it in ILC1.
Collapse
Affiliation(s)
- Remi Marrocco
- Center for Vaccine Innovation and Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Eduardo Lucero-Meza
- Center for Vaccine Innovation and Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Chris A Benedict
- Center for Vaccine Innovation and Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| |
Collapse
|
20
|
Kotagiri P, Rae WM, Bergamaschi L, Pombal D, Lee JY, Noor NM, Sojwal RS, Rubin SJS, Unger LW, Tolmeijer SH, Manferrari G, Bashford-Rogers RJM, Bingham DB, Stift A, Rogalla S, Gubatan J, Lee JC, Smith KGC, McKinney EF, Boyd SD, Lyons PA. Disease-specific B cell clones are shared between patients with Crohn's disease. Nat Commun 2025; 16:3689. [PMID: 40246842 PMCID: PMC12006383 DOI: 10.1038/s41467-025-58977-y] [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: 01/25/2023] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
B cells have important functions in gut homeostasis, and dysregulated B cell populations are frequently observed in patients with inflammatory bowel diseases, including both ulcerative colitis (UC) and Crohn's disease (CD). How these B cell perturbations contribute to disease remains largely unknown. Here, we perform deep sequencing of the B cell receptor (BCR) repertoire in four cohorts of patients with CD, together with healthy controls and patients with UC. We identify BCR clones that are shared between patients with CD but not found in healthy individuals nor in patients with UC, indicating CD-associated B cell immune responses. Shared clones are present in the inflamed gut mucosa, draining intestinal lymph nodes and blood, suggesting the presence of common CD-associated antigens that drive B cell responses in CD patients.
Collapse
Affiliation(s)
- Prasanti Kotagiri
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK.
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA.
| | - William M Rae
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Discovery Sciences, AstraZeneca, Cambridge Biomedical Campus, Cambridge, UK
| | - Laura Bergamaschi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Diana Pombal
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Ji-Yeun Lee
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Nurulamin M Noor
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Raoul S Sojwal
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, 94305, USA
| | - Samuel J S Rubin
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, 94305, USA
| | - Lukas W Unger
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Sofie H Tolmeijer
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Giulia Manferrari
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Rachael J M Bashford-Rogers
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Department of Biochemistry, South Parks Road, University of Oxford, Oxford, OX1 3QU, UK
| | - David B Bingham
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Anton Stift
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Stephan Rogalla
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, 94305, USA
| | - John Gubatan
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, 94305, USA
| | - James C Lee
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- The Francis Crick Institute and UCL Institute of Liver and Digestive Health, Division of Medicine, Royal Free Campus, London, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Eoin F McKinney
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Scott D Boyd
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Paul A Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK.
| |
Collapse
|
21
|
Nie J, Zhou L, Tian W, Liu X, Yang L, Yang X, Zhang Y, Wei S, Wang DW, Wei J. Deep insight into cytokine storm: from pathogenesis to treatment. Signal Transduct Target Ther 2025; 10:112. [PMID: 40234407 PMCID: PMC12000524 DOI: 10.1038/s41392-025-02178-y] [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: 08/09/2024] [Revised: 12/22/2024] [Accepted: 02/12/2025] [Indexed: 04/17/2025] Open
Abstract
Cytokine storm (CS) is a severe systemic inflammatory syndrome characterized by the excessive activation of immune cells and a significant increase in circulating levels of cytokines. This pathological process is implicated in the development of life-threatening conditions such as fulminant myocarditis (FM), acute respiratory distress syndrome (ARDS), primary or secondary hemophagocytic lymphohistiocytosis (HLH), cytokine release syndrome (CRS) associated with chimeric antigen receptor-modified T (CAR-T) therapy, and grade III to IV acute graft-versus-host disease following allogeneic hematopoietic stem cell transplantation. The significant involvement of the JAK-STAT pathway, Toll-like receptors, neutrophil extracellular traps, NLRP3 inflammasome, and other signaling pathways has been recognized in the pathogenesis of CS. Therapies targeting these pathways have been developed or are currently being investigated. While novel drugs have demonstrated promising therapeutic efficacy in mitigating CS, the overall mortality rate of CS resulting from underlying diseases remains high. In the clinical setting, the management of CS typically necessitates a multidisciplinary team strategy encompassing the removal of abnormal inflammatory or immune system activation, the preservation of vital organ function, the treatment of the underlying disease, and the provision of life supportive therapy. This review provides a comprehensive overview of the key signaling pathways and associated cytokines implicated in CS, elucidates the impact of dysregulated immune cell activation, and delineates the resultant organ injury associated with CS. In addition, we offer insights and current literature on the management of CS in cases of FM, ARDS, systemic inflammatory response syndrome, treatment-induced CRS, HLH, and other related conditions.
Collapse
Grants
- 82070217, 81873427 National Natural Science Foundation of China (National Science Foundation of China)
- 82100401 National Natural Science Foundation of China (National Science Foundation of China)
- 81772477, 81201848, 82473220 National Natural Science Foundation of China (National Science Foundation of China)
- 82330010,81630010,81790624 National Natural Science Foundation of China (National Science Foundation of China)
- National High Technology Research and Development Program of China, Grant number: 2021YFA1101500.
- The Hubei Provincial Natural Science Foundation (No.2024AFB050)
- Project of Shanxi Bethune Hospital, Grant Numbber: 2023xg02); Fundamental Research Program of Shanxi Province, Grant Numbber: 202303021211224
- The Key Scientific Research Project of COVID-19 Infection Emergency Treatment of Shanxi Bethune Hospital (2023xg01), 2023 COVID-19 Research Project of Shanxi Provincial Health Commission (No.2023XG001, No. 2023XG005), Four “Batches” Innovation Project of Invigorating Medical through Science and Technology of Shanxi Province (2023XM003), Cancer special Fund research project of Shanxi Bethune Hospital (No. 2020-ZL04), and External Expert Workshop Fund Program of Shanxi Provincial Health Commission(Proteomics Shanxi studio for Huanghe professor)
- Fundamental Research Program of Shanxi Province(No.202303021221192); 2023 COVID-19 Emergency Project of Shanxi Health Commission (Nos.2023XG001,2023XG005)
Collapse
Affiliation(s)
- Jiali Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Ling Zhou
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China
| | - Weiwei Tian
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xiansheng Liu
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Liping Yang
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xingcheng Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China.
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China.
| | - Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
22
|
John K, Huntress I, Smith E, Chou H, Tollison TS, Covarrubias S, Crisci E, Carpenter S, Peng X. Human long noncoding RNA VILMIR is induced by major respiratory viral infections and modulates the host interferon response. J Virol 2025; 99:e0014125. [PMID: 40130878 PMCID: PMC11998520 DOI: 10.1128/jvi.00141-25] [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: 02/04/2025] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
Long noncoding RNAs (lncRNAs) are a newer class of noncoding transcripts identified as key regulators of biological processes. Here, we aimed to identify novel lncRNA targets that play critical roles in major human respiratory viral infections by systematically mining large-scale transcriptomic data sets. Using bulk RNA-sequencing (RNA-seq) analysis, we identified a previously uncharacterized lncRNA, named virus-inducible lncRNA modulator of interferon response (VILMIR), that was consistently upregulated after in vitro influenza infection across multiple human epithelial cell lines and influenza A virus subtypes. VILMIR was also upregulated after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and respiratory syncytial virus (RSV) infections in vitro. We experimentally confirmed the response of VILMIR to influenza infection and interferon-beta (IFN-β) treatment in the A549 human epithelial cell line and found the expression of VILMIR was robustly induced by IFN-β treatment in a dose- and time-specific manner. Single-cell RNA-seq analysis of bronchoalveolar lavage fluid samples from coronavirus disease 2019 (COVID-19) patients uncovered that VILMIR was upregulated across various cell types, including at least five immune cells. The upregulation of VILMIR in immune cells was further confirmed in the human T cell and monocyte cell lines, SUP-T1 and THP-1, after IFN-β treatment. Finally, we found that knockdown of VILMIR expression reduced the magnitude of host transcriptional responses to both IFN-β treatment and influenza A virus infection in A549 cells. Together, our results show that VILMIR is a novel interferon-stimulated gene (ISG) that regulates the host interferon response and may be a potential therapeutic target for human respiratory viral infections upon further mechanistic investigation.IMPORTANCEIdentifying host factors that regulate the immune response to human respiratory viral infection is critical to developing new therapeutics. Human long noncoding RNAs (lncRNAs) have been found to play key regulatory roles during biological processes; however, the majority of lncRNA functions within the host antiviral response remain unknown. In this study, we identified that a previously uncharacterized lncRNA, virus-inducible lncRNA modulator of interferon response (VILMIR), is upregulated after major respiratory viral infections including influenza, severe acute respiratory syndrome coronavirus 2, and respiratory syncytial virus. We demonstrated that VILMIR is an interferon-stimulated gene that is upregulated after interferon-beta (IFN-β) in several human cell types. We also found that knockdown of VILMIR reduced the magnitude of host transcriptional responses to IFN-β treatment and influenza A infection in human epithelial cells. Our results reveal that VILMIR regulates the host interferon response and may present a new therapeutic target during human respiratory viral infections.
Collapse
Affiliation(s)
- Kristen John
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
- Genetics & Genomics Graduate Program, North Carolina State University, Raleigh, North Carolina, USA
| | - Ian Huntress
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
- Bioinformatics Graduate Program, North Carolina State University, Raleigh, North Carolina, USA
| | - Ethan Smith
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
- Bioinformatics Graduate Program, North Carolina State University, Raleigh, North Carolina, USA
| | - Hsuan Chou
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
| | - Tammy S. Tollison
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
| | - Sergio Covarrubias
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, USA
| | - Elisa Crisci
- Department of Population Health and Pathobiology, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
| | - Susan Carpenter
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, USA
| | - Xinxia Peng
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
- Bioinformatics Graduate Program, North Carolina State University, Raleigh, North Carolina, USA
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
23
|
Luo C, Ma C, Xu G, Lu C, Ma J, Huang Y, Nie L, Yu C, Xia Y, Liu Z, Zhu Y, Liu S. Hepatitis B surface antigen hijacks TANK-binding kinase 1 to suppress type I interferon and induce early autophagy. Cell Death Dis 2025; 16:304. [PMID: 40234418 PMCID: PMC12000394 DOI: 10.1038/s41419-025-07605-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/17/2025]
Abstract
There are close links between innate immunity and autophagy. However, the crosstalk between innate immunity and autophagy in host cells infected with hepatitis B virus (HBV) remains unclear. Here, we reported that HBsAg suppressed type I interferon production and induced the accumulation of autophagosomes. HBsAg boosted TANK-binding kinase 1 (TBK1) phosphorylation and depressed interferon regulatory factor 3 (IRF3) phosphorylation ex vivo and in vivo. Mechanistic studies showed that HBsAg interaction with the kinase domain (KD) of TBK1 augmented its dimerization but disrupted TBK1-IRF3 complexes. Using the TBK1 inhibitor, BX795, we discovered that HBsAg-enhanced TBK1 dimerization, promoting sequestosome-1 (p62) phosphorylation, was necessary for HBV-induced autophagy and HBV replication. Moreover, HBsAg blocked autophagosome-lysosome fusion by inhibiting the synaptosomal-associated protein 29 (SNAP29) promoter. Notably, liver tissues from HBsAg transgenic mice or chronic HBV patients revealed that IFNβ signaling was inhibited and incomplete autophagy was induced. These findings suggest a novel mechanism by which HBsAg targets TBK1 to inhibit type I interferon and induce early autophagy, possibly leading to persistent HBV infection. Molecular mechanisms of HBsAg suppression of the IFNβ signaling pathway and triggering of early autophagy. HBsAg targets the kinase domain of TBK1, thereby disrupting the TBK1-IRF3 complex and inhibiting type I interferon production. On the other hand, HBsAg enhances TBK1 dimerization and phosphorylation, which upregulates the phosphorylation of p62 to induce p62-mediated autophagy. Furthermore, HBV infection causes the accumulation of autophagosomes. This is achieved by HBsAg suppressing the SNAP29 promoter activity, which blocks autophagosome-lysosome fusion.
Collapse
Affiliation(s)
- Chuanjin Luo
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Caijiao Ma
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Gang Xu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Chengbo Lu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - June Ma
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Huang
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Longyu Nie
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Chen Yu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yongfang Xia
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhiqiang Liu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Ying Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China.
| | - Shi Liu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
24
|
Qi F, Yi Z, Liu Y, Jia D, Zhao H, Jiang G, Gong J. CMTM4 promotes PD-L1-mediated macrophage apoptosis by enhancing STAT2 phosphorylation in sepsis. Exp Cell Res 2025; 447:114519. [PMID: 40122504 DOI: 10.1016/j.yexcr.2025.114519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Macrophage apoptosis is a key contributor to the elimination of immune cells and increased susceptibility during sepsis. CKLF like MARVEL transmembrane domain containing 4 (CMTM4) is a membrane protein with four transmembrane domains. It has recently been implicated in the regulation of immune cell biological functions. However, its role in regulating macrophage apoptosis during sepsis has not been extensively studied. METHODS Clinical samples were analyzed to determine CMTM4 expression levels and their correlation with clinical examination results. An in vitro model was developed using C57BL/6 mice and the THP-1 cell line. An immunofluorescence analysis was used to assess protein expression levels, apoptosis, and protein co-localization. Western blotting (WB) was used to measure protein expression levels, while flow cytometry was used to detect cell apoptosis. Transcriptomic sequencing was conducted to identify differentially expressed genes and to perform a functional enrichment analysis. Transcription factors were screened using databases. Chromatin immunoprecipitation, followed by quantitative PCR (ChIP-qPCR), was conducted to analyze protein-DNA interactions, and co-immunoprecipitation (Co-IP) was used to examine protein-protein interactions. RESULTS CMTM4 expression in macrophages was upregulated in sepsis. The inhibition of CMTM4 expression reduced macrophage apoptosis. PD-L1 was identified as a key molecule regulated by CMTM4 in macrophage apoptosis. CMTM4 regulates PD-L1 by promoting the phosphorylation of its transcription factor, STAT2, rather than directly binding to PD-L1. CONCLUSION In sepsis, CMTM4 facilitates PD-L1-dependent macrophage apoptosis by enhancing STAT2 phosphorylation. This discovery offers new insights for the diagnosis and treatment of sepsis.
Collapse
Affiliation(s)
- Feng Qi
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhujun Yi
- Department of Hepatobiliary Surgery, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Yan Liu
- Department of Hepatobiliary Surgery, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Degong Jia
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hui Zhao
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Gang Jiang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Jianping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
25
|
Wang YQ, Wang S, Yi HM, Qian Y, Wang Y, Xu HM, Xu-Monette ZY, Au K, Tian S, Dong Y, Zhao J, Fu D, Mu RJ, Wang SY, Wang L, Young KH, Xu PP, Zhao WL. Practical microenvironment classification in diffuse large B cell lymphoma using digital pathology. Cell Rep Med 2025; 6:102030. [PMID: 40112808 PMCID: PMC12047489 DOI: 10.1016/j.xcrm.2025.102030] [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: 10/07/2024] [Revised: 01/15/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025]
Abstract
Diffuse large B cell lymphoma (DLBCL) is a heterogeneous B cell neoplasm with variable clinical outcomes influenced by both tumor-derived and lymphoma microenvironment (LME) alterations. A recent transcriptomic study identifies four DLBCL subtypes based on LME characteristics: germinal center (GC)-like, mesenchymal (MS), inflammatory (IN), and depleted (DP). However, integrating this classification into clinical practice remains challenging. Here, we utilize deconvolution methods to assess microenvironment component abundance, establishing an LME classification of DLBCL using immunohistochemistry markers and digital pathology based on CD3, CD8, CD68, PD-L1, and collagen. This staining-based algorithm demonstrates over 80% concordance with transcriptome-based classification. Single-cell sequencing confirms that the immune microenvironments distinguished by this algorithm align with transcriptomic profiles. Significant disparities in overall and progression-free survival are observed among LME subtypes following rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) or R-CHOP with targeted agents (R-CHOP-X) immunochemotherapy. LME subtypes differed from distinct immune escape mechanisms, highlighting specific immunotherapeutic targets and supporting application of this classification in future precision medicine trials.
Collapse
Affiliation(s)
- Yu-Qing Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuo Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong-Mei Yi
- Department of Pathology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Qian
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hai-Min Xu
- Department of Pathology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zijun Y Xu-Monette
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA
| | - Kelly Au
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Shuang Tian
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Dong
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Di Fu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong-Ji Mu
- Department of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu-Ye Wang
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Li Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China
| | - Ken H Young
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA.
| | - Peng-Peng Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Wei-Li Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China.
| |
Collapse
|
26
|
Wang J, Niu H, Kang J, Liu H, Dong X. Macrophage Polarization in Lung Diseases: From Mechanisms to Therapeutic Strategies. Immunol Invest 2025:1-27. [PMID: 40213814 DOI: 10.1080/08820139.2025.2490898] [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: 04/24/2025]
Abstract
Macrophages are pivotal immune cells involved in maintaining immune homeostasis and defending against pathogens. They exhibit significant plasticity and heterogeneity, enabling polarization into pro-inflammatory M1 or anti-inflammatory M2 phenotypes in response to distinct microenvironmental cues. The process of macrophage polarization is tightly regulated by complex signaling pathways and transcriptional networks. This review explores the factors influencing macrophage polarization, the associated signaling pathways, and their roles in the pathogenesis of lung diseases, including fibrosis, cancer, and chronic inflammatory conditions. By summarizing recent advances, we aim to provide insights into the immunoregulatory functions of macrophages and their therapeutic potential. Based on our review, it is believed that targeting macrophage polarization emerges as a promising approach for developing effective treatments for lung diseases, balancing inflammation and repair while mitigating disease progression.
Collapse
Affiliation(s)
- Jia Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Huajie Niu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Junwei Kang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Haiping Liu
- Department of Radiology, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, P.R. China
| | - Xiaoyang Dong
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, P.R. China
| |
Collapse
|
27
|
Cui Y, Zhang H, Deng Y, Fan O, Wang J, Xing Z, Tang J, Zhu W, Gong B, Sun YE. Shared and distinct peripheral blood immune cell landscape in MCTD, SLE, and pSS. Cell Biosci 2025; 15:42. [PMID: 40211396 PMCID: PMC11983850 DOI: 10.1186/s13578-025-01374-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/26/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Mixed connective tissue disease (MCTD) is a rare autoimmune disease, and little is known about its pathogenesis. Furthermore, MCTD, systemic lupus erythematosus (SLE), and primary Sjögren's syndrome (pSS) share many clinical, laboratory, and immunological manifestations. This overlap complicates early diagnosis and accurate treatment. METHODS The transcriptomic profiling of peripheral blood mononuclear cells (PBMCs) from MCTD patients was performed using both bulk RNA sequencing and single-cell RNA sequencing (scRNA-seq) for the first time. Additionally, we applied MCTD scRNA-seq data, along with datasets from SLE (GSE135779) and pSS (GSE157278) from the Gene Expression Omnibus database, to characterize and compare the similarities and heterogeneity among MCTD, SLE, and pSS. RESULTS We first resolved transcriptomic changes in peripheral blood immune cells of MCTD, and then revealed the shared and unique features among MCTD, SLE, and pSS. Analyses showed that the percentage of CD8+ effector T cells was increased, while mucosal-associated invariant T cells were decreased in all three diseases. Genes related to the 'interferon (IFN) γ response' and 'IFN α response' were significantly upregulated. SCENIC analysis revealed activation of STAT1 and IRF7 in disease states, targeting IFN-related genes. The IFN-II signaling network was notably elevated in all three diseases. Unique features of MCTD, SLE, and pSS were also identified. CONCLUSION We dissected the immune landscape of MCTD at single-cell resolution, providing new insights into the development of novel biomarkers and immunotherapies for MCTD. Furthermore, we offer insights into the transcriptomic similarities and heterogeneity across different autoimmune diseases, while highlighting prospective therapeutic targets.
Collapse
Affiliation(s)
- Yanling Cui
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huina Zhang
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yaxuan Deng
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Orion Fan
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Junbang Wang
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhonggang Xing
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jianping Tang
- Division of Rheumatology, Tongji Hospital of Tongji University School of Medicine, Shanghai, China.
| | - Wenmin Zhu
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Bangdong Gong
- Division of Rheumatology, Tongji Hospital of Tongji University School of Medicine, Shanghai, China.
| | - Yi Eve Sun
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China.
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
28
|
Huang HC, Fong M, Nowak I, Shcherbinina E, Lobo V, Besavilla DF, Huynh HT, Schön K, Westholm JO, Fernandez C, Patel AAH, Wiel C, Sayin VI, Anastasakis D, Angeletti D, Sarshad AA. Nuclear AGO2 supports influenza A virus replication through type-I interferon regulation. Nucleic Acids Res 2025; 53:gkaf268. [PMID: 40219968 PMCID: PMC11992678 DOI: 10.1093/nar/gkaf268] [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: 10/30/2024] [Revised: 03/03/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
The role of Argonaute (AGO) proteins and the RNA interference (RNAi) machinery in mammalian antiviral response has been debated. Therefore, we set out to investigate how mammalian RNAi impacts influenza A virus (IAV) infection. We reveal that IAV infection triggers nuclear accumulation of AGO2, which is directly facilitated by p53 activation. Mechanistically, we show that IAV induces nuclear AGO2 targeting of TRIM71and type-I interferon-pathway genes for silencing. Accordingly, Tp53-/- mice do not accumulate nuclear AGO2 and demonstrate decreased susceptibility to IAV infection. Hence, the RNAi machinery is highjacked by the virus to evade the immune system and support viral replication. Furthermore, the FDA-approved drug, arsenic trioxide, prevents p53 nuclear translocation, increases interferon response and decreases viral replication in vitro and in a mouse model in vivo. Our data indicate that targeting the AGO2:p53-mediated silencing of innate immunity may offer a promising strategy to mitigate viral infections.
Collapse
Affiliation(s)
- Hsiang-Chi Huang
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Michelle Fong
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Iwona Nowak
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Evgeniia Shcherbinina
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Vivian Lobo
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Danica F Besavilla
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Hang T Huynh
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Karin Schön
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Jakub O Westholm
- Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Stockholm University, Box 1031, SE-17121 Solna, Sweden
| | - Carola Fernandez
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Angana A H Patel
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Clotilde Wiel
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Volkan I Sayin
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Dimitrios G Anastasakis
- Department of Basic Sciences, School of Medicine, University of Crete, GR 70013 Heraklion ,Greece
| | - Davide Angeletti
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
- SciLifeLab, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Aishe A Sarshad
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
| |
Collapse
|
29
|
Li JX, Zhang J, Li CH, Zhang Q, Kong B, Wang PH. Human papillomavirus E2 proteins suppress innate antiviral signaling pathways. Front Immunol 2025; 16:1555629. [PMID: 40264759 PMCID: PMC12011818 DOI: 10.3389/fimmu.2025.1555629] [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: 01/05/2025] [Accepted: 03/17/2025] [Indexed: 04/24/2025] Open
Abstract
Human papillomavirus (HPV) is a major cause of cancers and benign lesions. High-risk (HR) types, including HPV16 and HPV18, are strongly implicated in cervical and other malignancies, while low-risk (LR) types, such as HPV11, are predominantly associated with benign conditions. Although the immune evasion of HPV oncoproteins E6 and E7 are extensively studied, the immunomodulatory functions of the E2 protein remain poorly underexplored. This study elucidates the role of HPV11 and HPV16 E2 proteins in modulating innate immune responses, focusing on their interaction with key innate antiviral signaling pathways. We demonstrate that HPV11 and HPV16 E2 proteins effectively suppress the activation of pivotal antiviral signaling pathways, including RIG-I/MDA5-MAVS, TLR3-TRIF, cGAS-STING, and JAK-STAT. Mechanistic analyses reveal that E2 proteins interact with the core components of type I interferon (IFN)-inducing pathways, inhibiting IRF3 phosphorylation and nuclear translocation, thereby attenuating IFN expression. Additionally, E2 disrupts the JAK-STAT signaling cascade by preventing the assembly of the ISGF3 complex, comprising STAT1, STAT2, and IRF9, ultimately inhibiting the transcription of interferon-stimulated genes (ISGs). These findings underscore the broader immunosuppressive role of HPV E2 proteins, complementing the well-established immune evasion mechanisms mediated by E6 and E7. This work advances our understanding of HPV-mediated immune evasion and positions the E2 protein as a promising target for therapeutic strategies aimed at augmenting antiviral immunity in HPV-associated diseases.
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
| | - 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
| | - Beihua 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
|
30
|
Park SJ, Garcia Diaz J, Comlekoglu T, Hahn YS. Type I IFN receptor blockade alleviates liver fibrosis through macrophage-derived STAT3 signaling. Front Immunol 2025; 16:1528382. [PMID: 40260261 PMCID: PMC12009845 DOI: 10.3389/fimmu.2025.1528382] [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: 11/14/2024] [Accepted: 03/11/2025] [Indexed: 04/23/2025] Open
Abstract
Liver macrophages play a role in the development of liver fibrosis progression via the regulation of inflammatory signaling. However, the precise mechanisms of macrophages contributing to liver fibrosis progression remain unclear. Using a preclinical model of CCl4-treated mice, we determined the composition of immune cells and the alteration of inflammatory gene expression. Our findings revealed a significant increase in liver macrophages, particularly those derived from infiltrating blood monocytes, in fibrotic mice. Moreover, the expression levels of type I IFN signature genes such as IFNα, IFNβ, ISG15, USP18, Ifi44, Ifit1, Ifit2, IRF3, and IRF7 were elevated in fibrotic mice. To determine the role of type I IFN signaling in liver fibrosis, we administered an IFNAR-1 antibody to block this pathway for 3 days prior to harvesting the liver. Notably, IFNAR-1 blockade reduced macrophage numbers compared to control mice and alleviated liver fibrosis in mice with increased hepatocyte proliferation and apoptosis. The ratio of P-STAT3/P-STAT1 in monocyte-derived macrophages was increased in the IFNAR-1 blockade group compared to fibrotic mice, and this was related to the appearance of M2 macrophage differentiation. Additionally, single-cell RNA-seq analysis indicated that IFNAR blockade affected inflammatory pathways involved in hepatocyte regeneration and fibrosis prevention. Taken together, IFNAR-1 blockade alleviates liver fibrosis progression by modulating macrophage inflammatory responses. These results provide insights for developing anti-fibrotic therapies against type I IFN signaling.
Collapse
Affiliation(s)
- Soo-Jeung Park
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Josefina Garcia Diaz
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Tina Comlekoglu
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Young S. Hahn
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
31
|
Martinez RJ, Hogquist KA. Sterile production of interferons in the thymus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf048. [PMID: 40184034 DOI: 10.1093/jimmun/vkaf048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/25/2025] [Indexed: 04/05/2025]
Abstract
T-cell central tolerance is controlled by thymocyte TCR recognition of self-peptides presented by thymic APCs. While thymic epithelial cells are essential for T-cell central tolerance, a variety of other traditional APCs also play critical roles in T-cell selection. Similar to how peripheral APCs require activation to become effective, thymic APCs also require activation to become tolerogenic. Recent studies have identified IFNs as an essential factor for the activation and generation of an optimally tolerogenic thymic environment. In this review, we focus on interferon (IFN) production within the thymus and its effects on thymic APCs and developing thymocytes. We also examine the importance of T-cell tolerance to IFN itself as well as to interferon-stimulated proteins generated during peripheral immune responses.
Collapse
Affiliation(s)
- Ryan J Martinez
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Kristin A Hogquist
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
32
|
Muhammad I, Contes K, Bility MT, Tang Q. Chasing Virus Replication and Infection: PAMP-PRR Interaction Drives Type I Interferon Production, Which in Turn Activates ISG Expression and ISGylation. Viruses 2025; 17:528. [PMID: 40284971 PMCID: PMC12031425 DOI: 10.3390/v17040528] [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: 03/09/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
The innate immune response, particularly the interferon-mediated pathway, serves as the first line of defense against viral infections. During virus infection, viral pathogen-associated molecular patterns (PAMPs) are recognized by host pattern recognition receptors (PRRs), triggering downstream signaling pathways. This leads to the activation of transcription factors like IRF3, IRF7, and NF-κB, which translocate to the nucleus and induce the production of type I interferons (IFN-α and IFN-β). Once secreted, type I interferons bind to their receptors (IFNARs) on the surfaces of infected and neighboring cells, activating the JAK-STAT pathway. This results in the formation of the ISGF3 complex (composed of STAT1, STAT2, and IRF9), which translocates to the nucleus and drives the expression of interferon-stimulated genes (ISGs). Some ISGs exert antiviral effects by directly or indirectly blocking infection and replication. Among these ISGs, ISG15 plays a crucial role in the ISGylation process, a ubiquitin-like modification that tags viral and host proteins, regulating immune responses and inhibiting viral replication. However, viruses have evolved counteractive strategies to evade ISG15-mediated immunity and ISGylation. This review first outlines the PAMP-PRR-induced pathways leading to the production of cytokines and ISGs, followed by a summary of ISGylation's role in antiviral defense and viral evasion mechanisms targeting ISG15 and ISGYlation.
Collapse
Affiliation(s)
| | | | | | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (I.M.); (K.C.); (M.T.B.)
| |
Collapse
|
33
|
Song Y, Lu J, Qin P, Chen H, Chen L. Interferon-I modulation and natural products: Unraveling mechanisms and therapeutic potential in severe COVID-19. Cytokine Growth Factor Rev 2025; 82:18-30. [PMID: 39261232 DOI: 10.1016/j.cytogfr.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 08/20/2024] [Indexed: 09/13/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to pose a significant global public health threat, particularly to older adults, pregnant women, and individuals with underlying chronic conditions. Dysregulated immune responses to SARS-CoV-2 infection are believed to contribute to the progression of COVID-19 in severe cases. Previous studies indicates that a deficiency in type I interferon (IFN-I) immunity accounts for approximately 15 %-20 % of patients with severe pneumonia caused by COVID-19, highlighting the potential therapeutic importance of modulating IFN-I signals. Natural products and their derivatives, due to their structural diversity and novel scaffolds, play a crucial role in drug discovery. Some of these natural products targeting IFN-I have demonstrated applications in infectious diseases and inflammatory conditions. However, the immunomodulatory potential of IFN-I in critical COVID-19 pneumonia and the natural compounds regulating the related signal pathway remain not fully understood. In this review, we offer a comprehensive assessment of the association between IFN-I and severe COVID-19, exploring its mechanisms and integrating information on natural compounds effective for IFN-I regulation. Focusing on the primary targets of IFN-I, we also summarize the regulatory mechanisms of natural products, their impact on IFNs, and their therapeutic roles in viral infections. Collectively, by synthesizing these findings, our goal is to provide a valuable reference for future research and to inspire innovative treatment strategies for COVID-19.
Collapse
Affiliation(s)
- Yuheng Song
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiani Lu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Pengcheng Qin
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Henan University, Kaifeng 475001, China
| | - Hongzhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 200032, China
| | - Lili Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
34
|
Tecle E, Warushavithana P, Li S, Blanchard MJ, Chhan CB, Bui T, Underwood RS, Bakowski MA, Troemel ER, Lažetić V. Conserved chromatin regulators control the transcriptional immune response to intracellular pathogens in Caenorhabditis elegans. PLoS Genet 2025; 21:e1011444. [PMID: 40193347 PMCID: PMC11975079 DOI: 10.1371/journal.pgen.1011444] [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: 09/30/2024] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
Robust transcriptional responses are critical for defense against infection. However, unrestrained immune responses can cause negative impacts such as damaging inflammation and slowed development. Here, we find that a class of transcriptional regulators previously associated with regulation of development in Caenorhabditis elegans, is also involved in repressing immune responses. Specifically, through forward genetics, we find that loss of lin-15B leads to constitutive expression of Intracellular Pathogen Response (IPR) genes. lin-15B encodes a transcriptional repressor with a conserved THAP domain that is associated with the DRM chromatin remodeling complex that regulates C. elegans development. We show that lin-15B mutants have increased resistance to natural intracellular pathogens, and the induction of IPR genes in lin-15B mutants relies on the MES-4 histone methyltransferase. We extend our analyses to other DRM and NuRD chromatin remodeling factors, as well as SUMOylation histone modifiers, showing that a broad range of chromatin-related factors can repress IPR gene expression. Altogether these findings suggest that conserved chromatin regulators may facilitate development in part by repressing damaging immune responses against intracellular pathogens.
Collapse
Affiliation(s)
- Eillen Tecle
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Paaramitha Warushavithana
- Department of Biological Sciences, Columbian College of Arts and Sciences, The George Washington University, District of Columbia,Washington, United States of America
| | - Samuel Li
- Department of Biological Sciences, Columbian College of Arts and Sciences, The George Washington University, District of Columbia,Washington, United States of America
| | - Michael J. Blanchard
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Crystal B. Chhan
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Theresa Bui
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Ryan S. Underwood
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Malina A. Bakowski
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Emily R. Troemel
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Vladimir Lažetić
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
- Department of Biological Sciences, Columbian College of Arts and Sciences, The George Washington University, District of Columbia,Washington, United States of America
| |
Collapse
|
35
|
Huang L, Zhong Q, Huang S, Yang K, Cai Y, Guo G. EBV enhances immunotherapy sensitivity in intrahepatic cholangiocarcinoma through cGAS-STING pathway activation. Hepatol Commun 2025; 9:e0674. [PMID: 40079734 PMCID: PMC11908760 DOI: 10.1097/hc9.0000000000000674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 01/16/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND The absence of representative Epstein-Barr virus-associated intrahepatic cholangiocarcinoma (EBVaICC) cell lines has limited our understanding of the molecular and immunological characteristics of this cancer subtype. METHODS We reviewed patients with metastatic cholangiocarcinoma at Sun Yat-sen University Cancer Center from January 2015 to August 2023. Among them, 22 patients with EBVaICC and 66 patients with non-EBVaICC who received anti-PD1 treatment were included. Additionally, 2 EBV-positive ICC cell lines, RBE-EBV and HuH28-EBV, were developed through cell-to-cell infection. Stable EBV infection and responsiveness to viral reactivation were confirmed. Transcriptomic and bioinformatics analyses were performed, and in vitro experiments examined the immune effects of EBV-positive ICC. Key immune-related genes and cytokines were validated by reverse transcription quantitative polymerase chain reaction and ELISA in cell lines and patient plasma samples. RESULTS In this study, we found that patients with EBVaICC showed enhanced immune responses and improved overall and progression-free survival compared to patients with non-EBVaICC. We first successfully established and validated 2 EBV-positive ICC cell lines (RBE-EBV and HuH28-EBV). These cell lines were confirmed for stable EBV infection and displayed responsiveness to viral reactivation, making them suitable for future studies. Transcriptomic analyses and in vitro studies revealed that EBV activated the cGAS-STING pathway, resulting in MHC-I upregulation and CXCL10 secretion in ICC cells, which collectively enhanced CD8+ T cell chemotaxis and cytotoxicity. Furthermore, ELISA analysis showed higher plasma levels of CXCL10 and IFN-γ in patients with EBVaICC, suggesting a potential role for EBV in enhancing immunotherapy sensitivity in this subtype. CONCLUSIONS The established EBV-positive ICC cell lines revealed enhanced immunogenicity driven by cGAS-STING pathway activation, providing valuable models for future research and insights into the mechanisms of improved immunotherapy sensitivity in EBVaICC.
Collapse
Affiliation(s)
- Lingli Huang
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Qian Zhong
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- State Key Laboratory of Oncology, Experimental Research Department, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Silan Huang
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Kejia Yang
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Yuchen Cai
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- State Key Laboratory of Oncology, Experimental Research Department, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Guifang Guo
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| |
Collapse
|
36
|
Huang B, Qu Y, Zhang H, Ma J, Chen J, Jie C, Liu F, Chu Z, Liu Y, Han Y, Wang X, Wang W. The characterization of a novel IRF8-like homolog and its role in the immune modulation of the sea urchin Strongylocentrotus intermedius. FISH & SHELLFISH IMMUNOLOGY 2025; 159:110179. [PMID: 39922248 DOI: 10.1016/j.fsi.2025.110179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/23/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Interferon regulatory factor (IRF) proteins, functioning as transcription factors, are essential for various animal species' innate immune defense and stress responses. However, further research is required to elucidate the roles of IRF in echinoderms. In this study, a new IRF gene (SiIRF8-like) was obtained from the sea urchin (Strongylocentrotus intermedius). The open reading frame for SiIRF8-like spanned 2004 bp and encoded a protein composed of 667 amino acids. Domain prediction analysis revealed a typical IRF domain at the N-terminus and an IRF3 domain at the C terminus of the SiIRF8-like protein, exhibiting similar amino acid sequences across different species. Phylogenetic analyses indicated that SiIRF8-like proteins were closely related to mollusk IRF8 proteins. Quantitative real-time PCR revealed detectable levels of SiIRF8-like mRNA in all sea urchin tissues examined, with the highest expression observed in coelomocytes. Furthermore, lipopolysaccharide and polyinosinic-polycytidylic acid treatments significantly increased transcript expression levels of SiIRF8-like. Subcellular localization experiments revealed that SiIRF8-like is mainly localized in the nucleus. Additionally, dual-luciferase reporter assays indicated that overexpression of SiIRF8-like in HEK293T cells could specifically activate reporter genes such as interleukin 6, interferon α/β/γ, activating protein 1, and interferon-stimulated response element. Finally, the overexpressed SiIRF8-like could promote the phosphorylation of protein kinases (JNK and Erk1/2). These preliminary findings regarding the immune functions linked to the SiIRF8-like protein offer valuable insights into the innate immunity mechanisms of invertebrate IRFs and provide theoretical support for developing disease-resistant strains of sea urchins.
Collapse
Affiliation(s)
- Baoyu Huang
- School of Fisheries, Ludong University, Yantai, China
| | - Yifan Qu
- School of Fisheries, Ludong University, Yantai, China
| | - Haikun Zhang
- School of Fisheries, Ludong University, Yantai, China
| | - Jilv Ma
- School of Fisheries, Ludong University, Yantai, China
| | - Jiwen Chen
- School of Fisheries, Ludong University, Yantai, China
| | - Cui Jie
- School of Fisheries, Ludong University, Yantai, China
| | - Fengchen Liu
- School of Fisheries, Ludong University, Yantai, China
| | - Zhongyi Chu
- School of Fisheries, Ludong University, Yantai, China
| | - Yaqiong Liu
- School of Fisheries, Ludong University, Yantai, China
| | - Yijing Han
- School of Fisheries, Ludong University, Yantai, China
| | - Xiaotong Wang
- School of Fisheries, Ludong University, Yantai, China.
| | - Wenhao Wang
- Yantai Marine Economic Research Institute, Yantai, China.
| |
Collapse
|
37
|
Chen PK, Wey SJ, Chen DY. Toward a personalized therapy of still's disease based on immunologic endotypes: a narrative review. Expert Rev Clin Immunol 2025; 21:401-412. [PMID: 39925174 DOI: 10.1080/1744666x.2025.2465406] [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: 10/15/2024] [Revised: 01/05/2025] [Accepted: 02/07/2025] [Indexed: 02/11/2025]
Abstract
INTRODUCTION Accumulative evidence indicates that both innate and adaptive immunity are involved in pathogenesis of Still's disease, an autoinflammatory disease. With Increasing insights into the pathogenesis of Still's disease coupled with the availability of emerging targeted therapeutics, it may be the unmet need for personalizing therapy and achieving a treat-to-target goal. We aim to summarize the available evidence regarding immunopathogenesis of Still's disease and therapeutic strategies based on immunologic endotypes. AREAS COVERED We searched MEDLINE database using the PubMed interface and reviewed relevant English-language literature from 1971 to 2024. This review focuses on the existing evidence on pathophysiology and immunological endotypes of Still's disease and their implications for personalized strategies for patients with this disease. EXPERT OPINION Targeting the complex immunopathogenesis of Still's disease, emerging new agents are available for treatment, including biologic disease-modifying anti-rheumatic drugs (bDMARDs) and targeted synthetic DMARDs (tsDMARDs) such as Janus kinase inhibitors (JAKi). According to the updated evidence, meta-analyses, and recommendations, we propose a flow chart emphasizing personalized therapeutic strategies based on immunological endotypes. Hopefully, the therapeutic strategy might help guide the optimal selection of b/tsDMARDs to achieve a 'treat-to-target' goal in Still's disease. This proposed flow chart will be updated as newer evidence emerges.
Collapse
Affiliation(s)
- Po-Ku Chen
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
- Translational Medicine Laboratory, Rheumatology and Immunology Center, Taichung, Taiwan
| | - Shiow-Jiuan Wey
- Division of Dermatology, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Der-Yuan Chen
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
- Translational Medicine Laboratory, Rheumatology and Immunology Center, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
38
|
Aljabali AAA, Obeid M, Gammoh O, El-Tanani M, Tambuwala MM. Guardians at the gate: Unraveling Type I interferon's role and challenges posed by anti-interferon antibodies in COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:135-169. [PMID: 40246343 DOI: 10.1016/bs.pmbts.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The intricate interplay involving Type I interferon (IFN), anti-interferon antibodies, and COVID-19 elucidates a complex symphony within the immune system. This chapter thoroughly explores the dynamic landscape of Type I IFN, delineating its pivotal role as the guardian of the immune response. As SARS-CoV-2 engages the host, the delicate balance of IFN induction and signaling pathways is disrupted, resulting in a nuanced impact on the severity and pathogenesis of COVID-19. Clinical studies illuminate a critical link between impaired IFN response and severe outcomes, uncovering genetic factors contributing to susceptibility. Furthermore, the emergence of anti-interferon antibodies proves to be a disruptive force, compromising the immune arsenal and correlating with disease severity. Our chapter encompasses diagnostic and prognostic implications, highlighting the importance of assays in identifying levels of IFN and anti-interferon antibodies. This chapter examines the possible incorporation of interferon-related biomarkers in COVID-19 diagnostics, offering predictive insights into disease progression. On the therapeutic front, efforts to manipulate the IFN pathway undergo scrutiny, encountering complexities in light of anti-interferon antibodies. This chapter concludes by outlining prospective avenues for precision medicine, emphasizing the imperative need for a comprehensive comprehension of the IFN landscape and its intricate interaction with COVID-19.
Collapse
Affiliation(s)
- Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan.
| | - Mohammad Obeid
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Omar Gammoh
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Mohamed El-Tanani
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, United Arab Emirates
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln, United Kingdom.
| |
Collapse
|
39
|
Huang Z, Gao C, Huang S, Lin S, Zhang W, You J, Chen X, Zhou P, Zhang G, Gong L. The alpha-coronavirus E protein inhibits the JAK-STAT pathway signaling by triggering STAT2 degradation through OPTN- and NBR1-mediated selective autophagy. Autophagy 2025:1-18. [PMID: 40091174 DOI: 10.1080/15548627.2025.2479671] [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: 12/05/2024] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/19/2025] Open
Abstract
The zoonotic transmission of coronaviruses continues to pose a considerable threat to humans. Swine acute diarrhea syndrome coronavirus (SADS-CoV), a bat coronavirus related to HKU2, causes severe economic losses in the pig industry and has the potential to trigger outbreaks in humans. However, our understanding of how SADS-CoV evades the host's innate immunity remains limited, hindering effective responses to potential human outbreaks. In this study, we demonstrate that the SADS-CoV envelope protein (E) inhibits type I interferon (IFN-I) signaling by inducing the degradation of STAT2 via the macroautophagy/autophagy-lysosome pathway. Mechanistically, the E protein evades host innate immunity by promoting STAT2 degradation through autophagy, mediated by the NBR1 and OPTN receptors. Notably, ubiquitination of E protein is required for the autophagic degradation of STAT2. Additionally, lysine residue K61 of the E protein is crucial for its stable expression; however, it is not involved in its ubiquitination. In conclusion, our study reveals a novel mechanism by which the E protein disrupts IFN-I signaling by targeting STAT2 via autophagy, enhancing our understanding of SADS-CoV's immune evasion strategies and providing potential drug targets for controlling viral infections.Abbreviations: 3-MA: 3-methyladenine; ATG: autophagy related; BafA1: bafilomycin A1; BSA: bovine serum albumin; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; CC: coiled-coil; CHX: cycloheximide; Co-IP: co-immunoprecipitation; DAPI: 4',6-diamidino-2-phenylindole; DBD: DNA-binding domain; DMEM: Dulbecco's Modified Eagle's medium; DMSO: dimethyl sulfoxide; E, Envelope. FW: four-tryptophan; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; HA: hemagglutinin; hpt: hours post-treatment; IF: indirect immunofluorescence; IFNB/IFN-β: interferon beta; IgG: immunoglobulin G; ISG: IFN-stimulated genes; ISRE: interferon-stimulated response element; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MOI: multiplicity of infection; NBR1: NBR1 autophagy cargo receptor; OPTN: optineurin; PBS: phosphate-buffered saline; PRRs: pattern recognition receptors; qPCR: quantitative polymerase chain reaction; SAR: selective autophagy receptor; SQSTM1/p62: sequestosome 1; STAT: signal transduction and activator of transcription; TBS-T: Tris-buffered saline with Tween 20; TCID50: 50% tissue culture infective dose; TOLLIP: toll interacting protein; Ub: ubiquitin; UBA: C-terminal ubiquitin-associated; VSV: vesicular stomatitis virus; WB: western blotting. WT: wild type.
Collapse
Affiliation(s)
- Zhao Huang
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Chenyang Gao
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Shaohong Huang
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Sizhan Lin
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - WenBo Zhang
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jianyi You
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiongnan Chen
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Pei Zhou
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Guihong Zhang
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Lang Gong
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| |
Collapse
|
40
|
Al-Janabi A, Martin P, Simpson C, Rhys H, Khan AR, Eyre S, Christofi M, Foulkes AC, Skelton A, Viatte S, Barton A, Morris AP, Smith CH, Griffiths CEM, Warren RB. Blood Single-Cell Transcriptomic and Proteomic Signatures of Paradoxical Eczema in Patients with Psoriasis Treated with Biologics. J Invest Dermatol 2025:S0022-202X(25)00370-7. [PMID: 40157420 DOI: 10.1016/j.jid.2025.02.153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 01/10/2025] [Accepted: 02/07/2025] [Indexed: 04/01/2025]
Abstract
Biologics targeting the TNF and IL-17/23 axis are highly effective treatments for psoriasis but can result in cutaneous adverse events. The pathogenesis of paradoxical eczema, the occurrence of an atopic dermatitis phenotype after biologic initiation in people with psoriasis, is unknown. Using single-cell RNA sequencing and mass cytometry, we found increased expression of TNF, IFN-γ, and IFN-α and their signaling pathways in paradoxical eczema case cell clusters compared with that in matched psoriasis controls. Genetic variants influencing the expression of chemokine signaling and TNF pathway genes were associated with paradoxical eczema in a separate genotyped cohort, and this association was independent of known atopic risk loci. This suggests that paradoxical eczema has a predominantly type 1 systemic inflammatory signature and that genetic susceptibility to aberrant chemokine and TNF pathway signaling could contribute to development of this phenotype during biologic treatment.
Collapse
Affiliation(s)
- Ali Al-Janabi
- Centre for Dermatology Research, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom.
| | - Paul Martin
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | | | | | | | - Steve Eyre
- Centre for Dermatology Research, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom; Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom
| | - Maria Christofi
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom
| | - Amy C Foulkes
- Centre for Dermatology Research, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | | | - Sebastien Viatte
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Anne Barton
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Catherine H Smith
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom; St. John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Christopher E M Griffiths
- Centre for Dermatology Research, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom; St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom; Department of Dermatology, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Richard B Warren
- Centre for Dermatology Research, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
41
|
Metwally H. STAT Signature Dish: Serving Immunity with a Side of Dietary Control. Biomolecules 2025; 15:487. [PMID: 40305224 PMCID: PMC12024614 DOI: 10.3390/biom15040487] [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: 02/17/2025] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
Immunity is a fundamental aspect of animal biology, defined as the host's ability to detect and defend against harmful pathogens and toxic substances to preserve homeostasis. However, immune defenses are metabolically demanding, requiring the efficient allocation of limited resources to balance immune function with other physiological and developmental needs. To achieve this balance, organisms have evolved sophisticated signaling networks that enable precise, context-specific responses to internal and external cues. These networks are essential for survival and adaptation in multicellular systems. Central to this regulatory architecture is the STAT (signal transducer and activator of Transcription) family, a group of versatile signaling molecules that govern a wide array of biological processes across eukaryotes. STAT signaling demonstrates remarkable plasticity, from orchestrating host defense mechanisms to regulating dietary metabolism. Despite its critical role, the cell-specific and context-dependent nuances of STAT signaling remain incompletely understood, highlighting a significant gap in our understanding. This review delves into emerging perspectives on immunity, presenting dynamic frameworks to explore the complexity and adaptability of STAT signaling and the underlying logic driving cellular decision-making. It emphasizes how STAT pathways integrate diverse physiological processes, from immune responses to dietary regulation, ultimately supporting organismal balance and homeostasis.
Collapse
Affiliation(s)
- Hozaifa Metwally
- Laboratory of Immune Regulation, The World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
42
|
Fang K, Yuan S, Zhang X, Zhang J, Sun SL, Li X. Regulation of immunogenic cell death and potential applications in cancer therapy. Front Immunol 2025; 16:1571212. [PMID: 40207233 PMCID: PMC11979251 DOI: 10.3389/fimmu.2025.1571212] [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: 02/05/2025] [Accepted: 03/11/2025] [Indexed: 04/11/2025] Open
Abstract
Immunogenic cell death (ICD), a type of regulatory cell death, plays an important role in activating the adaptive immune response. Activation of the tumor-specific immune response is accompanied by the cell surface exposure of calreticulin and heat-shock proteins, the secretion of adenosine triphosphate, and the release of high mobility group box-1. In this review, we summarize and classify the latest types of ICD inducers and their molecular mechanisms, and discuss the effects and potential applications of inducing ICD by chemotherapy drugs, targeted drugs, and oncolytic viruses in clinical research. We also explore the potential role of epigenetic modifiers in the induction of ICD, and clarify the synergistic anti-tumor effects of nano-pulse stimulation, radiosensitizers for radiotherapy, photosensitizers for photodynamic therapy, photothermal therapy, and other physical stimulation, combined with radiotherapy and chemotherapy induced-ICD, in multimodal immunotherapy. In addition, we elucidate the molecular mechanism of ICD in detail, including the calcium imbalance, mitochondrial stress, and the interactions in the tumor microenvironment. Ultimately, this review aims to offer deeper insight into the factors and mechanisms of ICD induction and provide a theoretical basis for the future development of ICD-based immunotherapy.
Collapse
Affiliation(s)
- Kun Fang
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Shuai Yuan
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Xue Zhang
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Jingdong Zhang
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
- Department of Medical Oncology, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
| | - Shu-lan Sun
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Xiaoxi Li
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| |
Collapse
|
43
|
Zheng H, Qiu C, Tian H, Zhu X, Yin B, Zhou Z, Li X, Zhao J. Host restriction factors against porcine epidemic diarrhea virus: a mini-review. Vet Res 2025; 56:67. [PMID: 40128890 PMCID: PMC11934732 DOI: 10.1186/s13567-025-01500-4] [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: 08/31/2024] [Accepted: 12/31/2024] [Indexed: 03/26/2025] Open
Abstract
Porcine epidemic diarrhea is an acute contagious disease caused by porcine epidemic diarrhea virus (PEDV), which severely constrains the development of the global swine industry. Host restriction factors constitute a vital defensive barrier against viral infections, typically interacting with viruses at specific stages of their replication process to disrupt it. Considering that traditional PEDV vaccines often struggle to effectively activate mucosal immunity in sows and thereby fail to provide reliable passive immunity to piglets via milk, this review focuses on the host restriction factors that play crucial roles in restricting PEDV infection and replication. The aim is to identify potential targets for the development of anti-PEDV drugs and offer insights for the exploration of novel vaccine adjuvants.
Collapse
Affiliation(s)
| | - Cunyi Qiu
- Gansu Polytechnic College of Animal Husbandry & Engineering, Wuwei, 733006, China
| | - Haolun Tian
- Northwest a&F University, Yangling, 712000, China
| | - Xiaofu Zhu
- Xianyang Polytechnic Institute, Xianyang, 712000, China
| | - Baoying Yin
- Xianyang Polytechnic Institute, Xianyang, 712000, China
| | - Zhiding Zhou
- Key Laboratory of Marine Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Xuezhao Li
- Gansu Polytechnic College of Animal Husbandry & Engineering, Wuwei, 733006, China
| | - Jingjing Zhao
- Department of Pharmaceutical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
44
|
Lang J, Bergner T, Zinngrebe J, Lepelley A, Vill K, Leiz S, Wlaschek M, Wagner M, Scharffetter-Kochanek K, Fischer-Posovszky P, Read C, Crow YJ, Hirschenberger M, Sparrer KMJ. Distinct pathogenic mutations in ARF1 allow dissection of its dual role in cGAS-STING signalling. EMBO Rep 2025:10.1038/s44319-025-00423-7. [PMID: 40128408 PMCID: PMC7617634 DOI: 10.1038/s44319-025-00423-7] [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: 08/15/2024] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/26/2025] Open
Abstract
Tight control of cGAS-STING-mediated DNA sensing is crucial to avoid auto-inflammation. The GTPase ADP-ribosylation factor 1 (ARF1) is crucial to maintain cGAS-STING homeostasis and various pathogenic ARF1 variants are associated with type I interferonopathies. Functional ARF1 inhibits STING activity by maintaining mitochondrial integrity and facilitating COPI-mediated retrograde STING trafficking and deactivation. Yet the factors governing the two distinct functions of ARF1 remained unexplored. Here, we dissect ARF1's dual role by a comparative analysis of disease-associated ARF1 variants and their impact on STING signalling. We identify a de novo heterozygous s.55 C > T/p.R19C ARF1 variant in a patient with type I interferonopathy symptoms. The GTPase-deficient variant ARF1 R19C selectively disrupts COPI binding and retrograde transport of STING, thereby prolonging innate immune activation without affecting mitochondrial integrity. Treatment of patient fibroblasts in vitro with the STING signalling inhibitors H-151 and amlexanox reduces chronic interferon signalling. Summarizing, our data reveal the molecular basis of a ARF1-associated type I interferonopathy allowing dissection of the two roles of ARF1, and suggest that pharmacological targeting of STING may alleviate ARF1-associated auto-inflammation.
Collapse
Affiliation(s)
- Johannes Lang
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Tim Bergner
- Central Facility for Electron Microscopy, Ulm University, Ulm, Germany
| | - Julia Zinngrebe
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Alice Lepelley
- Institut Imagine-Inserm UMR1163, Laboratory of Neurogenetics and Neuroinflammation, Université Paris Cité, Paris, France
| | - Katharina Vill
- Department of Pediatric Neurology and Developmental Medicine, Dr. von Hauner Children's Hospital, LMU-University of Munich, Munich, Germany
| | - Steffen Leiz
- Division of Neuropediatrics, Klinikum Dritter Orden, Munich, Germany
| | - Meinhard Wlaschek
- Department of Dermatology and Allergic Diseases, Ulm University Medical Center, Ulm, Germany
| | - Matias Wagner
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum Munich, Munich, Germany
| | | | - Pamela Fischer-Posovszky
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
- German Center for Child and Adolescent Health (DZKJ), Partner site Ulm, Ulm, Germany
| | - Clarissa Read
- Central Facility for Electron Microscopy, Ulm University, Ulm, Germany
| | - Yanick J Crow
- Institut Imagine-Inserm UMR1163, Laboratory of Neurogenetics and Neuroinflammation, Université Paris Cité, Paris, France
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | | | - Konstantin M J Sparrer
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany.
| |
Collapse
|
45
|
Icedo-Nuñez S, Luna-Ramirez RI, Enns RM, Speidel SE, Hernández J, Zeng X, Sánchez-Castro MA, Aguilar-Trejo CM, Luna-Nevárez G, López-González MC, Reyna-Granados JR, Luna-Nevárez P. Validation of Polymorphisms Associated with the Immune Response After Vaccination Against Porcine Reproductive and Respiratory Syndrome Virus in Yorkshire Gilts. Vet Sci 2025; 12:295. [PMID: 40284797 PMCID: PMC12031381 DOI: 10.3390/vetsci12040295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/29/2025] Open
Abstract
Porcine respiratory and reproductive syndrome is a viral disease that impacts the health and profitability of swine farms, largely due to significant variation in the vaccination response. The objective was to identify and validate molecular markers associated with the antibody response in gilts following vaccination against the PRRSV. The study included one hundred (n = 100) 6-month-old Yorkshire gilts that were negative for the PRRSV. Gilts were randomly assigned to one of two treatments, PRRS-vaccinated (n = 75) and control (n = 25) groups. Blood samples collected on day 21 were analyzed to evaluate the antibody response, as indicated by the sample-to-positive (S/P) ratio, to the PRRSV following vaccination. DNA was extracted and genotyped using a low-density chip containing 10,000 single nucleotide polymorphisms (SNPs). A genome-wide association study (GWAS) was conducted to identify candidate SNPs associated with the S/P ratio, which were validated in two independent gilt populations (n = 226). The SNPs rs707264998, rs708860811, and rs81358818 in the genes RNF144B, XKR9, and BMAL1, respectively, were significantly associated (p < 0.01) with the S/P ratio and demonstrated an additive effect. In conclusion, three SNPs are proposed as candidate markers for an enhanced immune response to vaccination against the PRRSV and may be beneficial in genetic selection programs.
Collapse
Affiliation(s)
- Salvador Icedo-Nuñez
- Departamento de Ciencias Agronómicas y Veterinarias, Instituto Tecnológico de Sonora, Ciudad Obregón 85000, Mexico
| | - Rosa I. Luna-Ramirez
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - R. Mark Enns
- Department of Animal Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Scott E. Speidel
- Department of Animal Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Jesús Hernández
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo A.C., Hermosillo 83304, Mexico
| | - Xi Zeng
- Zoetis Inc., VMRD Genetics R&D, Kalamazoo, MI 49007, USA
| | | | - Carlos M. Aguilar-Trejo
- Departamento de Ciencias Agronómicas y Veterinarias, Instituto Tecnológico de Sonora, Ciudad Obregón 85000, Mexico
| | - Guillermo Luna-Nevárez
- Departamento de Ciencias Agronómicas y Veterinarias, Instituto Tecnológico de Sonora, Ciudad Obregón 85000, Mexico
| | - Martha C. López-González
- Departamento de Ciencias Agronómicas y Veterinarias, Instituto Tecnológico de Sonora, Ciudad Obregón 85000, Mexico
| | - Javier R. Reyna-Granados
- Departamento de Ciencias Agronómicas y Veterinarias, Instituto Tecnológico de Sonora, Ciudad Obregón 85000, Mexico
| | - Pablo Luna-Nevárez
- Departamento de Ciencias Agronómicas y Veterinarias, Instituto Tecnológico de Sonora, Ciudad Obregón 85000, Mexico
| |
Collapse
|
46
|
Tang M, Shi J, Pang Y, Zhou S, Liu J, Wu C, Li M, Zeng X, Wang Q. Nailfold videocapillaroscopy findings are associated with IIM subtypes and IFN activation. Arthritis Res Ther 2025; 27:62. [PMID: 40114239 PMCID: PMC11924856 DOI: 10.1186/s13075-025-03532-9] [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: 11/21/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
OBJECTIVE This study aimed to characterize nailfold videocapillaroscopy (NVC) features in patients with different subtypes of Idiopathic inflammatory myopathy (IIM) and to investigate the correlations between NVC findings, myositis-specific antibody (MSA) subtypes, disease activity, cytokine profiles, and interferon-stimulated gene (ISG) expression levels. METHODS This cross-sectional observational single-center study included 55 IIM patients, categorized into MDA5 (+), anti-aminoacyl-tRNA-synthetase antibodies (ARS) (+), and MSA(-) groups based on their MSA profiles. Demographic data, laboratory tests, and NVC assessments were systematically collected and analyzed. The relative expression of type I ISGs in whole blood, as well as serum cytokine and chemokine profiles, were measured. Statistical analyses were performed to explore correlations between NVC scores and clinical parameters, including serum biomarkers. RESULTS NVC abnormalities were observed in most IIM patients, with significant differences in NVC features among the MSA subgroups. The MDA5(+) group exhibited significantly higher scores for capillary dilation (P < 0.01), giant capillaries (P < 0.05), microhemorrhages (P < 0.01), and abnormal capillary morphology (P < 0.05) compared to the ARS (+) group. ISG expression and cytokine levels were upregulated in IIM patients, with active disease patients showing significantly higher levels of certain ISGs and cytokines compared to clinically stable patients. Notably, specific NVC score dimensions were positively correlated with the levels of certain ISGs and cytokines. For example, microhemorrhage, capillary dilation, and capillary density all had significantly positive correlations with MX1, IFI27, IP-10, RANTES, and GROα (P < 0.05). And giant capillary is also related to levels of IFI27, SDF-1α, IP-10, RANTES, and GROα (P < 0.05). CONCLUSION IIM patients exhibit distinct NVC abnormalities, which vary across different MSA subtypes. NVC findings have potential clinical value in screening disease activity and interferon pathway activation in IIM patients.
Collapse
Affiliation(s)
- Mingwei Tang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Ministry of Education, Peking Union Medical College Hospital, Peking Union Medical College Hospital, Beijing, 100730, China
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Jia Shi
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Ministry of Education, Peking Union Medical College Hospital, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Yiyun Pang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Ministry of Education, Peking Union Medical College Hospital, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Shuang Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Ministry of Education, Peking Union Medical College Hospital, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Jinjing Liu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Ministry of Education, Peking Union Medical College Hospital, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Chanyuan Wu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Ministry of Education, Peking Union Medical College Hospital, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Ministry of Education, Peking Union Medical College Hospital, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Ministry of Education, Peking Union Medical College Hospital, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Qian Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases Key Laboratory of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences, Ministry of Education, Peking Union Medical College Hospital, Peking Union Medical College Hospital, Beijing, 100730, China.
| |
Collapse
|
47
|
Wang C, Huang Y, Li L, Huang X, Huang Y, Fang X, Long Y. Antiviral Therapy-Induced Changes in Long Non-Coding RNA Expression Profiles in Umbilical Cord Blood and Placental Tissues of Hepatitis B Virus-Infected Pregnant Women. Int J Womens Health 2025; 17:835-844. [PMID: 40123756 PMCID: PMC11927581 DOI: 10.2147/ijwh.s511524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/08/2025] [Indexed: 03/25/2025] Open
Abstract
Background Hepatitis B virus (HBV) is a major global health concern, with maternal-fetal transmission being the primary route of transmission, which can lead to chronic HBV infection in newborns. Long non-coding RNAs (lncRNAs) play crucial roles in gene regulation and immune responses, but their involvement in HBV transmission during pregnancy remains unclear. This study aimed to assess the impact of tenofovir disoproxil fumarate (TDF)-based antiviral therapy on lncRNA expression profiles and immune signaling pathways in umbilical cord blood and placental tissues and to identify potential therapeutic targets for preventing intrauterine HBV infection. Materials and Methods Umbilical cord serum and placental tissues were collected from six HBV carriers. Three carriers received TDF-based antiviral therapy, and the remaining carriers who did not receive antiviral therapy served as controls. LncRNA microarray analysis and bioinformatics were used to evaluate the effects of antiviral therapy on lncRNA expression profiles and signaling pathways. Results Antiviral therapy exerted minimal effects on lncRNA expression profiles in umbilical cord blood. In placental tissues, significant alterations in lncRNA expression profiles were observed, including 249 upregulated and 381 downregulated lncRNAs. Antiviral therapy activated innate immune pathways, such as intracellular DNA sensing, chemokine signaling, type I interferon, Jak-Stat, and interferon-γ-mediated adaptive immunity. Through intersection analysis, CPED1 was found differentially expressed in both cord blood and placental tissues. KEGG pathway analysis suggested that low CPED1 expression may inhibit virus transmission via the JAK-STAT pathway. Conclusion This study demonstrated that TDF-based antiviral therapy altered lncRNA expression and activated immune signaling pathways in placental tissues, offering insights into the molecular mechanisms of maternal-fetal HBV transmission.
Collapse
Affiliation(s)
- Cuimin Wang
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Department of Obstetrics & Gynecology, Guangxi Zhuang Autonomous Region People’s Hospital, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Yuting Huang
- Department of Obstetrics & Gynecology, Youjiang Medical College for Nationalities, Baise City, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Lanfeng Li
- Department of Obstetrics & Gynecology, Guangxi Zhuang Autonomous Region People’s Hospital, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Xizhen Huang
- Department of Obstetrics & Gynecology, Guangxi Zhuang Autonomous Region People’s Hospital, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Yin Huang
- Department of Obstetrics & Gynecology, Guangxi Zhuang Autonomous Region People’s Hospital, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Xiang Fang
- Department of Obstetrics & Gynecology, Guangxi Zhuang Autonomous Region People’s Hospital, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Yu Long
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Medical Simulator Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| |
Collapse
|
48
|
Huang M, Jin Y, Zhao D, Liu X. Potential role of lactylation in intrinsic immune pathways in lung cancer. Front Pharmacol 2025; 16:1533493. [PMID: 40166469 PMCID: PMC11955616 DOI: 10.3389/fphar.2025.1533493] [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: 11/24/2024] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
Lung cancer, one of the most lethal malignancies, has seen its therapeutic strategies become a focal point of significant scientific attention. Intrinsic immune signaling pathways play crucial roles in anti-tumor immunity but face clinical application challenges despite promising preclinical outcomes. Lactylation, an emerging research focus, may influences lung cancer progression by modulating the functions of histones and non-histone proteins. Recent findings have suggested that lactylation regulates key intrinsic immune molecules, including cGAS-STING, TLR, and RIG-I, thereby impacting interferon expression. However, the precise mechanisms by which lactylation governs intrinsic immune signaling in lung cancer remain unclear. This review presents a comprehensive and systematic analysis of the relationship between lactylation and intrinsic immune signaling pathways in lung cancer and emphasizes the innovative perspective of linking lactylation-mediated epigenetic modifications with immune regulation. By thoroughly examining current research findings, this review uncovers potential regulatory mechanisms and highlights the therapeutic implications of targeting lactylation in lung cancer. Future investigations into the intricate interactions between lactylation and intrinsic immunity are anticipated to unveil novel therapeutic targets and strategies, potentially improving patient survival outcomes.
Collapse
Affiliation(s)
- Mengdie Huang
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ye Jin
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Dandan Zhao
- Department of Thoracic Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xingren Liu
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
49
|
Shan Y, Pang H, Tang Y, Yang N, Wang R, Yang F, Qin B. Altered LY6E and TRIM6 expression in PBMCs correlated with HBsAg clearance and response to Peg-IFN-α treatment in HBeAg-negative chronic hepatitis B patients. Virol J 2025; 22:74. [PMID: 40089754 PMCID: PMC11909810 DOI: 10.1186/s12985-025-02689-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/01/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Pegylated interferon alpha (Peg-IFN-α) has the potential to eradicate hepatitis B surface antigen (HBsAg). This study aimed to investigate whether the expression levels of lymphocyte antigen 6 family member E (LY6E) and tripartite motif-containing protein 6 (TRIM6) mRNAs in peripheral blood mononuclear cells (PBMCs) of hepatitis B e antigen (HBeAg)-negative chronic hepatitis B virus (HBV) patients is associated with the response to Peg-IFN-α treatment and HBsAg clearance. METHODS In this prospective study, HBeAg-negative chronic HBV patients treated with Peg-IFN-α were followed for 48 weeks. The participants were classified into two groups, the virological response (VR) group and nonvirological response (NVR) group, according to the changes in HBV DNA and HBsAg levels observed at week 48 of treatment. Furthermore, these patients were divided into a serological response (SR) group and a nonserological response (NSR) group, depending on whether they exhibited a loss of serum HBsAg or evidence of seroconversion. The expression levels of LY6E and TRIM6 mRNAs in PBMCs were evaluated using real-time quantitative PCR with fluorescence detection. The diagnostic performance of LY6E and TRIM6 was assessed by analyzing the receiver operating characteristic (ROC) curve and calculating the area under the ROC curve (AUC). RESULTS After the treatment period, the observed VR and SR rates were 44.64% and 28.57%, respectively. Dynamic changes in LY6E and TRIM6 mRNA levels were significantly different between the VR and NVR groups and between the SR and NSR groups. Multivariate analysis revealed that TRIM6 was independently associated with VR at weeks 12 and 24 of Peg-IFN-α therapy and with SR at week 12; in addition, LY6E was independently associated with VR at week 12 and SR at week 24. At week 24, the area under the curve (AUC) for LY6E in the prediction of VR was 0.6942, and the AUC for the prediction of SR was 0.7766; at week 12, TRIM6 had AUCs of 0.7600 for the prediction of VR and 0.8469 for the prediction of SR. CONCLUSIONS LY6E and TRIM6 are important biomarkers for early therapeutic responses to Peg-IFN-α and HBsAg clearance. TRIAL REGISTRATION Registration number: 2023 - 311. Date of registration: 1 October 2023.
Collapse
MESH Headings
- Humans
- Hepatitis B, Chronic/drug therapy
- Hepatitis B, Chronic/virology
- Hepatitis B, Chronic/blood
- Male
- Interferon-alpha/therapeutic use
- Female
- Leukocytes, Mononuclear/metabolism
- Leukocytes, Mononuclear/virology
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/drug effects
- Adult
- Antiviral Agents/therapeutic use
- Hepatitis B Surface Antigens/blood
- Prospective Studies
- Middle Aged
- Tripartite Motif Proteins/genetics
- Tripartite Motif Proteins/metabolism
- Hepatitis B e Antigens/blood
- DNA, Viral/blood
- DNA, Viral/genetics
- Treatment Outcome
- Ubiquitin-Protein Ligases/genetics
- GPI-Linked Proteins/genetics
- Hepatitis B virus/genetics
- Hepatitis B virus/drug effects
- Young Adult
- Viral Load/drug effects
- Antigens, Surface
Collapse
Grants
- KJQN202100401, to Fan Yang Scientific and Technological Research Program of Chongqing Municipal Education Commission
- KJQN202100401, to Fan Yang Scientific and Technological Research Program of Chongqing Municipal Education Commission
- KJQN202100401, to Fan Yang Scientific and Technological Research Program of Chongqing Municipal Education Commission
- 2022-02-016-Y, to Yiru Shan Scientific and Technological Program of Basic Research and Achievement Transformation in Jiulongpo District of Chongqing
- cstc2024ycjh-bgzxm0095, to Bo Qin the Chongqing Talents Program
- cstc2024ycjh-bgzxm0095, to Bo Qin the Chongqing Talents Program
- cstc2024ycjh-bgzxm0095, to Bo Qin the Chongqing Talents Program
- cstc2024ycjh-bgzxm0095, to Bo Qin the Chongqing Talents Program
- CSTC2020JCYJ-MSXMX0221, to Bo Qin the Chongqing Natural Science Foundation of China
- CSTC2020JCYJ-MSXMX0221, to Bo Qin the Chongqing Natural Science Foundation of China
- CSTC2020JCYJ-MSXMX0221, to Bo Qin the Chongqing Natural Science Foundation of China
- W0101, to Fan Yang Program for Youth Innovation in Future Medicine, Chongqing Medical University
- W0101, to Fan Yang Program for Youth Innovation in Future Medicine, Chongqing Medical University
- W0101, to Fan Yang Program for Youth Innovation in Future Medicine, Chongqing Medical University
Collapse
Affiliation(s)
- Yiru Shan
- Department of Infectious Diseases, Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Pang
- Department of Infectious Diseases, Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yao Tang
- Department of Infectious Diseases, Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Na Yang
- Central Laboratory, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Rui Wang
- Central Laboratory, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fan Yang
- Department of Infectious Diseases, Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Bo Qin
- Department of Infectious Diseases, Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
50
|
Chivatá-Ávila JA, Rojas-Estevez P, Muñoz-Suarez AM, Caro-Morales E, Rengifo AC, Torres-Fernández O, Lozano JM, Álvarez-Díaz DA. Mild Zika Virus Infection in Mice Without Motor Impairments Induces Working Memory Deficits, Anxiety-like Behaviors, and Dysregulation of Immunity and Synaptic Vesicle Pathways. Viruses 2025; 17:405. [PMID: 40143332 PMCID: PMC11946058 DOI: 10.3390/v17030405] [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: 01/30/2025] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND The Zika virus (ZIKV) is an arbovirus linked to "Congenital Zika Syndrome" and a range of neurodevelopmental disorders (NDDs), with microcephaly as the most severe manifestation. Milder NDDs, such as autism spectrum disorders and delays in neuropsychomotor and language development, often go unnoticed in neonates, resulting in long-term social and academic difficulties. Murine models of ZIKV infection can be used to mimic part of the spectrum of motor and cognitive deficits observed in humans. These can be evaluated through behavioral tests, enabling comparison with gene expression profiles and aiding in the characterization of ZIKV-induced NDDs. OBJECTIVES This study aimed to identify genes associated with behavioral changes following a subtle ZIKV infection in juvenile BALB/c mice. METHODS Neonatal mice were subcutaneously inoculated with ZIKV (MH544701.2) on postnatal day 1 (DPN) at a dose of 6.8 × 103 PFU. Viral presence in the cerebellum and cortex was quantified at 10- and 30-days post-infection (DPI) using RT-qPCR. Neurobehavioral deficits were assessed at 30 DPI through T-maze, rotarod, and open field tests. Next-Generation Sequencing (NGS) was performed to identify differentially expressed genes (DEGs), which were analyzed through Gene Ontology (GO) and KEGG enrichment. Gene interaction networks were then constructed to explore gene interactions in the most enriched biological categories. RESULTS A ZIKV infection model was successfully established, enabling brain infection while allowing survival beyond 30 DPI. The infection induced mild cognitive behavioral changes, though motor and motivational functions remained unaffected. These cognitive changes were linked to the functional repression of synaptic vesicles and alterations in neuronal structure, suggesting potential disruptions in neuronal plasticity. CONCLUSIONS Moderate ZIKV infection with circulating strains from the 2016 epidemic may cause dysregulation of genes related to immune response, alterations in cytoskeletal organization, and modifications in cellular transport mediated by vesicles. Despite viral control, neurocognitive effects persisted, including memory deficits and anxiety-like behaviors, highlighting the long-term neurological consequences of ZIKV infection in models that show no apparent malformations.
Collapse
Affiliation(s)
- Jaime Alexander Chivatá-Ávila
- Grupo de Genómica de Microorganismos Emergentes, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud, Bogotá 111321, Colombia; (J.A.C.-Á.); (P.R.-E.)
| | - Paola Rojas-Estevez
- Grupo de Genómica de Microorganismos Emergentes, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud, Bogotá 111321, Colombia; (J.A.C.-Á.); (P.R.-E.)
| | - Alejandra M. Muñoz-Suarez
- Grupo de Animales de Laboratorio, Dirección de Producción, Instituto Nacional de Salud, Bogotá 111321, Colombia; (A.M.M.-S.); (E.C.-M.)
| | - Esthefanny Caro-Morales
- Grupo de Animales de Laboratorio, Dirección de Producción, Instituto Nacional de Salud, Bogotá 111321, Colombia; (A.M.M.-S.); (E.C.-M.)
| | - Aura Caterine Rengifo
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud, Bogotá 111321, Colombia; (A.C.R.); (O.T.-F.)
| | - Orlando Torres-Fernández
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud, Bogotá 111321, Colombia; (A.C.R.); (O.T.-F.)
| | - Jose Manuel Lozano
- Grupo Mimetismo Molecular de los Agentes Infecciosos, Departamento de Farmacia, Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá 11001, Colombia;
| | - Diego A. Álvarez-Díaz
- Grupo de Genómica de Microorganismos Emergentes, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud, Bogotá 111321, Colombia; (J.A.C.-Á.); (P.R.-E.)
- Grupo de Investigación y Desarrollo en Vacunas y Biológicos Estratégicos en Salud Pública, Dirección de Producción, Instituto Nacional de Salud, Bogotá 111321, Colombia
| |
Collapse
|