1
|
Yin J, Wang S, Zhang Z, Ge J, Zhang Q, Sun Y, Yin X, Wang X. The rabies virus matrix protein (RABV M) interacts with host histone deacetylase 6 (HDAC6) to activate the MEK/ ERK signaling pathway and enhance viral replication. Vet Microbiol 2025; 305:110537. [PMID: 40300412 DOI: 10.1016/j.vetmic.2025.110537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/01/2025]
Abstract
Rabies virus (RABV) is the causative agent of rabies, posing a severe threat to human and animal health. The matrix (M) protein of RABV plays crucial roles during viral infection. In this study, we identified RABV M protein interacted with host histone deacetylase 6 (HDAC6) through a combination of immunoprecipitation and mass spectrometry analysis. Specifically, the catalytic domains of HDAC6 (amino acids 435-835) was shown to be critical for the interaction between HDAC6 and the RABV M protein. Overexpression of HDAC6 significantly enhanced RABV replication, whereas inhibition of HDAC6 expression or its deacetylase activity had the opposite effect,indicating that HDAC6 is a positive regulator of RABV replication. We further determined that RABV infection actives the MEK/ERK pathway, and inhibition of this pathway with U0126 significantly reduced viral titers. Moreover, HDAC6 positively regulated MEK/ERK pathway activation in a manner independent of its deacetylase activity but dependent on the presence of HDAC6 during virus infection. Finally, we demonstrated that co-expression of RABV M enhanced the role of HDAC6 in facilitating MEK/ERK pathway activation. Collectively, our findings demonstrate that RABV exploits the HDAC6-M interaction to hijack the MEK/ERK signaling axis, which is essential for viral replication. Notably, HDAC6 facilitates MEK/ERK activation in a deacetylase activity-independent manner, revealing a novel mechanism by which viruses manipulate host machinery. These results highlight HDAC6 as a potential therapeutic target for combating rabies.
Collapse
Affiliation(s)
- Juanbin Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China; College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Shasha Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China
| | - Zhixiong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Junwei Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Qiang Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China
| | - Yuefeng Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China
| | - Xiangping Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China.
| | - Xiangwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China.
| |
Collapse
|
2
|
Tan S, Chen Y, Wang J, Li Z, Li J, Liu H, Yu J, Yue R, Xiao J, Wu H, Yan J, Zou J, Feng H. Black carp OTUD1 negatively regulates antiviral innate immunity via deubiquitination and degradation of IRF3/7. FISH & SHELLFISH IMMUNOLOGY 2025; 163:110407. [PMID: 40350101 DOI: 10.1016/j.fsi.2025.110407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
IRF3 and IRF7 function importantly in type I interferon production. Ovarian tumor deubiquitinase 1 (OTUD1) is a critical member of OUT deubiquitinases, which has been identified as a negative regulator in IRF3-mediated interferon signaling in mammals. However, the role of teleost OTUD1 in the immunity remains unclear. In this study, black carp (Mylopharyngodon piceus) OTUD1 (bcOTUD1) was cloned and identified, distributed in the nucleus and cytoplasm, and involved in virus-induced innate immune response. Overexpression of bcOTUD1 inhibits bcIRF3/7-mediated interferon production and antiviral ability, while knockdown bcOTUD1 promotes host antiviral capacity. In addition, bcOTUD1 could degrade bcIRF3/7 protein levels, while proteasome inhibitor MG132, lysosome inhibitor chloroquine and autophagy inhibitor 3-MA are able to restore this degradation. Co-immunoprecipitation assay has revealed that bcOTUD1 interacts with bcIRF3/7, and removes K63-linked ubiquitination of bcIRF3 and K48- and K63-linked ubiquitination of bcIRF7. To conclude, our research has brought to light for the first time in teleost that OTUD1 degrade IRF3/7 through decreasing their ubiquitination, thereby restraining IRF3/7-mediated antiviral immune response, which provides an important reference for the study of teleost interferon signaling.
Collapse
Affiliation(s)
- Shasha Tan
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Yixin Chen
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jiaxin Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Zhan'ao Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jinyi Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Hanbo Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jiajie Yu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Ruimin Yue
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jun Xiao
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Hui Wu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jun Yan
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China.
| | - Jun Zou
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Hao Feng
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
3
|
Cuenca-Escalona J, Sweep MWD, Gorris MAJ, Duiveman-de Boer T, Cambi A, Flórez-Grau G, de Vries JM. Distinct roles of PGE2 signaling via EP2 and EP4 in circulating pDCs: Implications for immune modulation in the tumor microenvironment. J Leukoc Biol 2025; 117:qiaf034. [PMID: 40116121 DOI: 10.1093/jleuko/qiaf034] [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: 08/06/2024] [Revised: 12/30/2024] [Accepted: 03/19/2025] [Indexed: 03/23/2025] Open
Abstract
Dendritic cells (DCs) play a pivotal role in orchestrating adaptive immunity in response to environmental cues such as prostaglandin E2 (PGE2). Tumors are known to establish a microenvironment rich in PGE2. Tumor-derived PGE2 is regarded as mediator of regulatory features in DCs, facilitating immune evasion and tumor progression. In DCs, the effects of PGE2 are mediated through the E-prostanoid receptor type 2 (EP2) and EP4. While the immunomodulatory effects of PGE2 signaling via EP2/4 in monocyte-derived DCs (moDCs) is well established, its role in human blood plasmacytoid DCs (pDCs) is poorly characterized. Therefore, in this study we investigated the effect of EP2 and EP4 signaling on pDC function, as well as the relevance of modulating these receptors in pDCs exposed to tumor-derived PGE2. Our findings reveal that EP2 and EP4 exhibit distinct functions in pDCs. PGE2-EP4 signaling mediates the upregulation of maturation markers (e.g., CD83 and HLA-DR), enhances a CCR7-based migratory function, impairs the production of proinflammatory mediators (e.g., interferon α and CXCL9), and stimulates the expansion of CD8 T cells with a marked suppressive phenotype. In contrast, PGE2-EP2 signaling hinders the upregulation of maturation markers and induces the expansion of CD8 T cells with a suppressive character. Additionally, using different in vitro tumor models, we show that EP2/4 blockade modulates the phenotype of pDCs exposed to tumor-derived PGE2. Together, these results identify the distinctive role of EP2 and EP4 signaling in pDCs and illustrate the potential therapeutic benefit of targeting this signaling axis to mitigate tumor-induced pDCs dysfunction.
Collapse
Affiliation(s)
- Jorge Cuenca-Escalona
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 26-28 6500HB, Nijmegen, the Netherlands
| | - Mark W D Sweep
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 26-28 6500HB, Nijmegen, the Netherlands
- Department of Medical Oncology, Radboud University Medical Center, Geert Grooteplein Zuid 10 6525GA, Nijmegen, the Netherlands
| | - Mark A J Gorris
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 26-28 6500HB, Nijmegen, the Netherlands
| | - Tjitske Duiveman-de Boer
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 26-28 6500HB, Nijmegen, the Netherlands
| | - Alessandra Cambi
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 26-28 6500HB, Nijmegen, the Netherlands
| | - Georgina Flórez-Grau
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 26-28 6500HB, Nijmegen, the Netherlands
| | - Jolanda M de Vries
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 26-28 6500HB, Nijmegen, the Netherlands
| |
Collapse
|
4
|
Froom ZSCS, Callaghan NI, Davenport Huyer L. Cellular crosstalk in fibrosis: insights into macrophage and fibroblast dynamics. J Biol Chem 2025:110203. [PMID: 40334985 DOI: 10.1016/j.jbc.2025.110203] [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: 03/13/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
Pathological fibrosis, the excessive deposition of extracellular matrix and tissue stiffening that causes progressive organ dysfunction, underlies diverse chronic diseases. The fibrotic microenvironment is driven by the dynamic microenvironmental interaction between various cell types; macrophages and fibroblasts play central roles in fibrotic disease initiation, maintenance, and progression. Macrophage functional plasticity to microenvironmental stimuli modulates fibroblast functionality by releasing pro-inflammatory cytokines, growth factors, and matrix remodeling enzymes that promote fibroblast proliferation, activation, and differentiation into myofibroblasts. Activated fibroblasts and myofibroblasts serve as the fibrotic effector cells, secreting extracellular matrix components and initiating microenvironmental contracture. Fibroblasts also modulate macrophage function through the release of their own pro-inflammatory cytokines and growth factors, creating bidirectional crosstalk that reinforces the chronic fibrotic cycle. The intricate interplay between macrophages and fibroblasts, including their secretomes and signaling interactions, leads to tissue damage and pathological loss of tissue function. In this review, we examine macrophage-fibroblast reciprocal dynamic interactions in pathological fibrotic conditions. We discuss the specific lineages and functionality of macrophages and fibroblasts implicated in fibrotic progression, with focus on their signal transduction pathways and secretory signalling that enables their pro-fibrotic behaviour. We then finish with a set of recommendations for future experimentation with the goal of developing a set of potential targets for anti-fibrotic therapeutic candidates. Understanding the cellular interactions between macrophages and fibroblasts provides valuable insights into potential therapeutic strategies to mitigate fibrotic disease progression.
Collapse
Affiliation(s)
- Zachary S C S Froom
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Neal I Callaghan
- Department of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Locke Davenport Huyer
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS B3H 4R2, Canada; Nova Scotia Health, Halifax, NS B3S 0H6, Canada.
| |
Collapse
|
5
|
Wang M, Chen Y, Xu B, Zhu X, Mou J, Xie J, Che Z, Zuo L, Li J, Jia H, Yu B. Recent advances in the roles of extracellular vesicles in cardiovascular diseases: pathophysiological mechanisms, biomarkers, and cell-free therapeutic strategy. Mol Med 2025; 31:169. [PMID: 40325357 PMCID: PMC12051314 DOI: 10.1186/s10020-025-01200-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 04/04/2025] [Indexed: 05/07/2025] Open
Abstract
Cardiovascular diseases (CVDs) represent a profound challenge with inflammation playing a significant role in their pathophysiology. Extracellular vesicles (EVs), which are membranous structures encapsulated by a lipid bilayer, are essential for intercellular communication by facilitating the transport of specific bioactive molecules, including microRNAs, proteins, and lipids. Emerging evidence suggests that the regulatory mechanisms governing cardiac resident cells are influenced by EVs, which function as messengers in intercellular communication and thereby contribute to the advancement of CVDs. In this review, we discuss the multifaceted biological functions of EVs and their involvement in the pathogenesis of various CVDs, encompassing myocardial infarction, ischemia-reperfusion injury, heart failure, atherosclerosis, myocarditis, cardiomyopathy, and aneurysm. Furthermore, we summarize the recent advancements in utilizing EVs as non-invasive biomarkers and in cell-free therapy based on EVs for the diagnosis and treatment of CVDs. Future research should investigate effective techniques for the isolation and purification of EVs from body fluids, while also exploring the pathways for the clinical translation of therapy based on EVs. Additionally, it is imperative to identify appropriate EV-miRNA profiles or combinations present in the circulation of patients, which could serve as biomarkers to improve the diagnostic accuracy of CVDs. By synthesizing and integrating recent research findings, this review aims to provide innovative perspectives for the pathogenesis of CVDs and potential therapeutic strategies.
Collapse
Affiliation(s)
- Mengyang Wang
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, 150001, People's Republic of China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Yuwu Chen
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, 150001, People's Republic of China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Biyi Xu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, 150001, People's Republic of China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Xinxin Zhu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, 150001, People's Republic of China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Junke Mou
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, 150001, People's Republic of China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Jiani Xie
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, 150001, People's Republic of China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Ziao Che
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, 150001, People's Republic of China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Liyang Zuo
- College of Pharmacy, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Ji Li
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, 150001, People's Republic of China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Haibo Jia
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, 150001, People's Republic of China.
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China.
| | - Bo Yu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, 150001, People's Republic of China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| |
Collapse
|
6
|
Steffen TL, Stafford JD, Bocke CR, Samson WK, Yosten GLC. The anorexigenic peptide nesfatin-1 dampens the B cell response to receptor-mediated stimulation through inhibition of NF-κB signaling. Am J Physiol Regul Integr Comp Physiol 2025; 328:R601-R610. [PMID: 40135734 DOI: 10.1152/ajpregu.00233.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/15/2024] [Accepted: 03/17/2025] [Indexed: 03/27/2025]
Abstract
Nesfatin-1, a posttranslational product of the protein encoded by the nucleobindin 2 gene (NUCB2), was functionally identified as an appetite regulatory molecule in rat hypothalamic nuclei. In the years following the discovery, those findings have been corroborated and expanded upon, and we now know that nesfatin-1 is expressed throughout peripheral tissues and exerts physiological effects beyond feeding control. Literature indicates that adipose tissue is one of the peripheral sources of NUCB2/nesfatin-1, and in this setting, it has anti-inflammatory effects that have recently been implicated in regulating chronic inflammation associated with diet-induced obesity. Currently, there are gaps in our understanding of what cell types within the adipose tissue compartment respond to nesfatin-1, in addition to the cellular mechanism(s) of this peptide. In this study, we sought to determine a mechanism by which this peptide might directly interact with the immune system starting with a human B cell line, Raji. We show that nesfatin-1 inhibits lipopolysaccharide (LPS) and B cell receptor (BCR) dual stimulation-mediated B cell growth, stimulation-induced cell death, and secretion of inflammatory mediators. Specifically, there was a reduced fold-change in B cell growth during stimulation which is paired with a reduction in the formation of apoptotic (annexin V+) cells. In addition, nesfatin-1 significantly reduced IgM secretion and modestly reduced TNFα secretion by stimulated B cells. The anti-inflammatory effects of nesfatin-1 overall are likely due to attenuation of NF-κB signaling, via inhibition of IκB degradation, in stimulated B cells.NEW & NOTEWORTHY This study establishes an interaction of nesfatin-1 and a human B cell line, Raji. Nesfatin-1 was shown to limit the B cell response to receptor-mediated stimulation, an action that has potential implications within the immune system and the development of chronic inflammation associated with the obese state. This study, along with previously published works, highlights a need for further research on nesfatin-1's interactions with adipocytes and immune cells.
Collapse
Affiliation(s)
- Tara L Steffen
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, United States
| | - Joshua D Stafford
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, United States
| | - Colleen R Bocke
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, United States
| | - Willis K Samson
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, United States
| | - Gina L C Yosten
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, United States
| |
Collapse
|
7
|
Zapico D, Espinosa J, Criado M, Gutiérrez D, Ferreras MDC, Benavides J, Pérez V, Fernández M. Immunohistochemical expression of TLR1, TLR2, TLR4, and TLR9 in the different types of lesions associated with bovine paratuberculosis. Vet Pathol 2025; 62:305-318. [PMID: 39720873 DOI: 10.1177/03009858241302850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
The factors that determine the appearance of the different pathologic forms associated with bovine paratuberculosis are not fully understood, but new research suggests a critical role of innate immunity. Toll-like receptors (TLRs) trigger the recognition of invading pathogens by innate immune cells and the onset of specific immune responses. The aim of this work was to assess, immunohistochemically, the expression of TLR1, TLR2, TLR4, and TLR9 in intestinal samples of 20 cows showing different types of paratuberculous lesions: uninfected controls, focal lesions, paucibacillary, and multibacillary diffuse forms. The majority of labeled cells were morphologically consistent with macrophages. A differential cell count was performed in the intestinal lamina propria, gut-associated lymphoid tissue, and mesenteric lymph node. TLR9 immunolabeling between the different types of lesions was compared using a complete H-score. Focal and diffuse paucibacillary forms contained significantly increased TLR2-expressing macrophages outside of the lesions compared with the controls and diffuse multibacillary forms, and moderate TLR9 immunolabeling within granulomas. In the multibacillary granulomatous lesions, the expression of TLR1 and TLR4 was observed as well as increased TLR9 expression compared with the rest of the groups. Differences in the predominance of one type or another of TLR allows us to elucidate the importance of the innate immune response and its possible role in the development of the different types of paratuberculosis lesions.
Collapse
Affiliation(s)
- David Zapico
- Universidad de León, León, Spain
- Instituto de Ganadería de Montaña, León, Spain
| | - José Espinosa
- Universidad de León, León, Spain
- Instituto de Ganadería de Montaña, León, Spain
| | - Miguel Criado
- Universidad de León, León, Spain
- Instituto de Ganadería de Montaña, León, Spain
| | - Daniel Gutiérrez
- Universidad de León, León, Spain
- Instituto de Ganadería de Montaña, León, Spain
| | | | | | - Valentín Pérez
- Universidad de León, León, Spain
- Instituto de Ganadería de Montaña, León, Spain
| | | |
Collapse
|
8
|
Xu X, Song H, Wu H, Zhang L, Lin F, Chen C, Zhang X, Liu Y, Li C, Fu Q. Effects of Environmentally Friendly Aquaculture Chamber Coatings on Enzyme Activities, Histology, and Transcriptome in the Liver of Larimichthys crocea. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2025; 27:78. [PMID: 40293578 DOI: 10.1007/s10126-025-10453-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/26/2025] [Indexed: 04/30/2025]
Abstract
Aquaculture vessels have emerged as a sustainable alternative to traditional offshore aquaculture. However, the biological impacts of protective coatings used for vessel interiors are still poorly understood. This study assessed acute stress responses of Larimichthys crocea to epoxy-based aquaculture coatings using actual culture (1-fold) and high-exposure (80-fold) concentrations. Liver analyses included antioxidant enzymes, histopathology, and transcriptomics over 12-96 h. Firstly, the effect of the 80-fold concentration group on the activities of catalase (CAT), superoxide dismutase (SOD), and peroxidase (POD) was more significant in the liver of L. crocea compared to the 1-fold concentration group. Similarly, histological observations revealed that the 80-fold concentration group produced more significant pathological changes in the liver than the 1-fold concentration group, including hepatocyte damage and vacuolization. Subsequently, through high-throughput sequencing, a total of 714.02 million clean reads were obtained, with 693.71 million of these reads successfully mapped onto the reference genome of L. crocea, identifying 13,709 differentially expressed genes (DEGs). KEGG pathway enrichment analysis showed that many DEGs following coating-treated were involved in protein processing in endoplasmic reticulum, oxidative phosphorylation, cytokine-cytokine receptor interaction, FoxO signaling pathway, and toll-like receptor signaling pathway. Finally, fifteen DEGs were selected for quantitative real-time PCR (qRT-PCR) analysis, and the results showed a significant correlation with RNA-seq results, verifying the reliability and accuracy of the high-throughput sequencing data. This study preliminarily revealed the stress responses induced by aquaculture vessel coatings in L. crocea and provided fundamental data into the scientific use of coatings on aquaculture vessels.
Collapse
Affiliation(s)
- Xuan Xu
- Qingdao Conson Oceantec Valley Development Co., Ltd, Qingdao, 266237, China
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Huayu Song
- Qingdao Conson Oceantec Valley Development Co., Ltd, Qingdao, 266237, China.
| | - Huicai Wu
- Qingdao Conson Oceantec Valley Development Co., Ltd, Qingdao, 266237, China
| | - Lu Zhang
- Qingdao Conson Oceantec Valley Development Co., Ltd, Qingdao, 266237, China
| | - Fengjun Lin
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Chonghui Chen
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Xiaoxu Zhang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Yiying Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Chao Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Qiang Fu
- Qingdao Conson Oceantec Valley Development Co., Ltd, Qingdao, 266237, China.
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
9
|
Park TJ, Choi BM, Hong H, Park JS, Kim SY. Anti-Inflammatory Effects of Ajuga decumbens Extract under Blue Light Stimulation. J Microbiol Biotechnol 2025; 35:e2502002. [PMID: 40295199 DOI: 10.4014/jmb.2502.02002] [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: 02/04/2025] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 04/30/2025]
Abstract
We investigated the potential of Ajuga decumbens Thunb. as an anti-inflammatory agent by utilizing plant resources to develop materials through the application of tissue culture and light-emitting diode (LED) cultivation technologies. To compare the changes between A. decumbens callus extract (ADCB) cultivated under blue monochromatic LED light and ADC (the negative control) cultivated under dark conditions, their morphological characteristics were tested and LC/MS analyses were conducted. ADCB exhibited a greenish hue compared with ADC and contained increased levels of specific compounds. The anti-inflammatory activities of the two samples were evaluated using LPS-stimulated macrophages. None of the samples exhibited cytotoxicity at any tested concentration. However, ADCB demonstrated a greater ability to reduce nitric oxide and key pro-inflammatory cytokines including interleukin-1β, interleukin-6, and tumor necrosis factor-α compared to the control. Furthermore, ADCB effectively suppressed the expression of inducible nitric oxide synthase and cyclooxygenase-2. It inhibited the phosphorylation of mitogen-activated protein kinase family proteins, including extracellular signal-regulated kinase, c-Jun N-terminal kinase, and p38, in a concentration-dependent manner. Tissue culture and LED cultivation technologies are significant methods for addressing plant supply challenges and enhancing the content of bioactive compounds, thereby increasing the applicability of plant materials. Moreover, ADCB produced using these technologies exhibited anti-inflammatory activity without causing irritation to human skin at active concentrations, suggesting its potential as a novel anti-inflammatory material.
Collapse
Affiliation(s)
- Tae-Jin Park
- Department of Pharmaceutical Engineering & Biotechnology, Sunmoon University, Asan 31460, Republic of Korea
| | - Byeong Min Choi
- Department of Pharmaceutical Engineering & Biotechnology, Sunmoon University, Asan 31460, Republic of Korea
| | - Hyehyun Hong
- Department of Pharmaceutical Engineering & Biotechnology, Sunmoon University, Asan 31460, Republic of Korea
| | - Jin-Soo Park
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Seung-Young Kim
- Department of Pharmaceutical Engineering & Biotechnology, Sunmoon University, Asan 31460, Republic of Korea
| |
Collapse
|
10
|
Sun H, Xu C, Xiong Z, Liu M, Ning X, Zhuang Y. Therapeutic prospects and potential mechanisms of Prdx6: as a novel target in musculoskeletal disorders. Front Physiol 2025; 16:1524100. [PMID: 40313876 PMCID: PMC12043587 DOI: 10.3389/fphys.2025.1524100] [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: 11/14/2024] [Accepted: 03/27/2025] [Indexed: 05/03/2025] Open
Abstract
With the global population aging, musculoskeletal disorders (MSDs) have posed significant physical and psychological health challenges for patients as well as a substantial economic burden on society. The advancements in conservative and surgical interventions for MSDs have been remarkable in recent years; however, the current treatment modalities still fall short of meeting the optimal requirements of patients. Recently, peroxiredoxin 6 (Prdx6) has gained considerable attention from researchers due to its remarkable antioxidative, anti-inflammatory, and anti-apoptotic properties. It has been found that Prdx6 is involved in multiple system diseases, including MSDs; however, the exact role of Prdx6 in MSDs is still lacking. This study aimed to summarize the structure, regulatory mechanism, and potential function of Prdx6. These findings may demonstrate Prdx6 as a novel target for inhibiting the advancement of MSDs.
Collapse
Affiliation(s)
- Hong Sun
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Chao Xu
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Zhilin Xiong
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Miao Liu
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xu Ning
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yong Zhuang
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
11
|
Jalali P, Shahmoradi A, Samii A, Mazloomnejad R, Hatamnejad MR, Saeed A, Namdar A, Salehi Z. The role of autophagy in cancer: from molecular mechanism to therapeutic window. Front Immunol 2025; 16:1528230. [PMID: 40248706 PMCID: PMC12003146 DOI: 10.3389/fimmu.2025.1528230] [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: 11/14/2024] [Accepted: 03/12/2025] [Indexed: 04/19/2025] Open
Abstract
Autophagy is a cellular degradation process that plays a crucial role in maintaining metabolic homeostasis under conditions of stress or nutrient deprivation. This process involves sequestering, breaking down, and recycling intracellular components such as proteins, organelles, and cytoplasmic materials. Autophagy also serves as a mechanism for eliminating pathogens and engulfing apoptotic cells. In the absence of stress, baseline autophagy activity is essential for degrading damaged cellular components and recycling nutrients to maintain cellular vitality. The relationship between autophagy and cancer is well-established; however, the biphasic nature of autophagy, acting as either a tumor growth inhibitor or promoter, has raised concerns regarding the regulation of tumorigenesis without inadvertently activating harmful aspects of autophagy. Consequently, elucidating the mechanisms by which autophagy contributes to cancer pathogenesis and the factors determining its pro- or anti-tumor effects is vital for devising effective therapeutic strategies. Furthermore, precision medicine approaches that tailor interventions to individual patients may enhance the efficacy of autophagy-related cancer treatments. To this end, interventions aimed at modulating the fate of tumor cells by controlling or inducing autophagy substrates necessitate meticulous monitoring of these mediators' functions within the tumor microenvironment to make informed decisions regarding their activation or inactivation. This review provides an updated perspective on the roles of autophagy in cancer, and discusses the potential challenges associated with autophagy-related cancer treatment. The article also highlights currently available strategies and identifies questions that require further investigation in the future.
Collapse
Affiliation(s)
- Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arvin Shahmoradi
- Department of Laboratory Medicine, Faculty of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Amir Samii
- Department of Hematology and Blood Transfusion, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Radman Mazloomnejad
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Hatamnejad
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Afshin Namdar
- Program in Cell Biology, The Hospital for Sick Children Peter Gilgan Centre for Research and Learning, Toronto, ON, United States
| | - Zahra Salehi
- Department of Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Guo X, Sen S, Gonzalez J, Hoffmann A. Macrophages maintain signaling fidelity in response to ligand mixtures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.645368. [PMID: 40236137 PMCID: PMC11996307 DOI: 10.1101/2025.03.27.645368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
As immune sentinel cells, macrophages are required to respond specifically to diverse immune threats and initiate appropriate immune responses. This stimulus-response specificity (SRS) is in part encoded in the signaling dynamics of the NFκB transcription factor. While experimental stimulus-response studies have typically focused on single defined ligands, in physiological contexts cells are exposed to multi-ligand mixtures. It remains unclear how macrophages combine multi-ligand information and whether they are able to maintain SRS in such complex exposure conditions. Here, we leveraged an established mathematical model that captures the heterogeneous single-cell NFκB responses of macrophage populations to extend experimental studies with systematic simulations of complex mixtures containing up to five ligands. Live-cell microscopy experiments for some conditions validated model predictions but revealed a discrepancy when TLR3 and TLR9 are stimulated. Refining the model suggested that the observed but unexpected ligand antagonism arises from a limited capacity for endosomal transport which is required for responses to CpG and pIC. With the updated model, we systematically analyzed SRS across all combinatorial-ligand conditions and employed three ways of quantifying SRS involving trajectory decomposition into informative trajectory features or machine learning. Our findings show that macrophages most effectively distinguish single-ligand stimuli, and distinguishability declines as more ligands are combined. However, even in complex combinatorial conditions, macrophages still maintain statistically significant distinguishability. These results indicate a robustness of innate immune response specificity: even in the context of complex exposure conditions, the NFκB temporal signaling code of macrophages can still classify immune threats to direct an appropriate response. Significance ≤120 Macrophages sense diverse pathogens within complex environments and respond appropriately. Experimental studies have found that the NFκB pathway responds with stimulus-specific dynamics when macrophages are exposed to single ligand stimuli. However, it remains unclear complex contexts might erode this stimulus-specificity. Here we systematically studies NFκB responses using a mathematical model that provides simulations of the heterogeneous population of single cell responses. We show that although the model is parameterized to single ligand data it can predict the responses to multi-ligand mixtures. Indeed, model validation uncovered signaling antagonism between two ligands and the underlying mechanism. Importantly, we found that NFκB signaling dynamics distinguish ligands within multi-ligand mixtures indicating a robustness of the NFκB temporal code that was not previously appreciated.
Collapse
|
13
|
Navarro León AI, Alonso-Hearn M, Muñoz M, Iglesias N, Badia-Bringué G, Iglesias T, Balseiro A, Casais R. Early Growth Response Factor 4 (EGR4) Expression in Gut Tissues and Regional Lymph Nodes of Cattle with Different Types of Paratuberculosis-Associated Lesions: Potential Role of EGR4 in Resilience to Paratuberculosis. Animals (Basel) 2025; 15:1012. [PMID: 40218405 PMCID: PMC11988129 DOI: 10.3390/ani15071012] [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: 02/18/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
Summary-data-based Mendelian randomization (SMR) analysis identified a novel cis-expression quantitative loci (cis-eQTL) associated with the upregulation of the expression of the early growth response factor 4 (EGR4) gene in animals with paratuberculosis (PTB)-associated multifocal lesions, which has been suggested to be modulating the NF-kβ-induced proinflammatory immune response to Mycobacterium avium subsp. paratuberculosis (Map) infection. To confirm these findings and to study the role of EGR4 expression in PTB resilience, the number of EGR4-expressing cells were analysed in paraffin-fixed gut tissues and regional lymph nodes of naturally Map-infected Holstein Friesian cows with focal, multifocal (subclinical and clinical), and diffuse lesions (intermediate and multibacillary), and in controls without lesions by quantitative anti-EGR4 immunohistochemistry. Subclinical animals with multifocal lesions showed a significantly higher number of EGR4-positive cells and were sacrificed at a significantly older average age than the remaining groups (p < 0.001 in all cases). We hypothesize that EGR4 could be mitigating the negative impact of Map infection on host clinical status through its involvement in three molecular mechanisms that promote resilience: (i) limiting NF-kβ-mediated proinflammatory responses, (ii) controlling tissue damage, acting as a brake on T-cell proliferation and cytokine production, and (iii) favouring tissue repair through interaction with epidermal growth factor receptor (EGFR).
Collapse
Affiliation(s)
- Alejandra Isabel Navarro León
- Center for Animal Biotechnology, Servicio Regional de Investigación y Desarrollo Agroalimentario (SERIDA), 33394 Deva, Spain; (A.I.N.L.); (M.M.)
| | - Marta Alonso-Hearn
- Animal Health Department, NEIKER, Basque Institute for Agricultural Research and Development, Basque Research and Technology Alliance (BRTA), 48160 Alava, Spain; (M.A.-H.); (G.B.-B.)
| | - Marta Muñoz
- Center for Animal Biotechnology, Servicio Regional de Investigación y Desarrollo Agroalimentario (SERIDA), 33394 Deva, Spain; (A.I.N.L.); (M.M.)
| | - Natalia Iglesias
- Center for Animal Biotechnology, Servicio Regional de Investigación y Desarrollo Agroalimentario (SERIDA), 33394 Deva, Spain; (A.I.N.L.); (M.M.)
| | - Gerard Badia-Bringué
- Animal Health Department, NEIKER, Basque Institute for Agricultural Research and Development, Basque Research and Technology Alliance (BRTA), 48160 Alava, Spain; (M.A.-H.); (G.B.-B.)
| | - Tania Iglesias
- Unidad de Consultoría Estadística, Servicios Científico-Técnicos, Universidad de Oviedo, Campus de Gijón, 33203 Gijón, Spain
| | - Ana Balseiro
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, 24071 León, Spain
- Instituto de Ganadería de Montaña (IGM, CSIC-ULE), 24346 León, Spain
| | - Rosa Casais
- Center for Animal Biotechnology, Servicio Regional de Investigación y Desarrollo Agroalimentario (SERIDA), 33394 Deva, Spain; (A.I.N.L.); (M.M.)
| |
Collapse
|
14
|
Sayed Y, Hassan M, Salem HM, Al-Amry K, Eid G. Probiotics/prebiotics effect on chicken gut microbiota and immunity in relation to heat-stress and climate-change mitigation. J Therm Biol 2025; 129:104097. [PMID: 40186955 DOI: 10.1016/j.jtherbio.2025.104097] [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: 09/18/2024] [Revised: 02/07/2025] [Accepted: 03/14/2025] [Indexed: 04/07/2025]
Abstract
Heat stress is a serious hazard that threatens world poultry production. The avian gut microbiome plays a critical role in improving nutrient utilization, competing with pathogens, stimulating an immune response, and reducing inflammatory reactions. Hence, the gut microbiome has a positive impact on the host's health which appears in the shape of improved body weight, feed conversion rate, and increased birds' productivity (meat or eggs). Accordingly, this review shed light on the chicken gut microbiome, its correlation with the immunity of chicken, and how this affects the general health condition of the bird as well as, the role of prebiotics and probiotics in improving the gut health and increasing birds' productivity, especially under climate change and heat stress condition. The review aims to focus on the significance of maintaining healthy chickens in order to increase the production of poultry meat to satisfy human needs. A robust microbiota and a well-functioning immune system synergistically contribute to the optimal health and productivity of chickens.
Collapse
Affiliation(s)
- Yara Sayed
- Department of Microbiology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
| | - Mariam Hassan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Galala University, New Galala City, 43511, Suez, Egypt; Department of Microbiology and Immunology, Faculty of Pharmacy Cairo University, Kasr El-Aini Street, Cairo, 11562, Cairo, Egypt.
| | - Heba M Salem
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt; Department of Diseases of Birds, Rabbits, Fish & their Care & Wildlife, School of Veterinary Medicine, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Khaled Al-Amry
- Department of Microbiology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Gamal Eid
- Department of Microbiology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| |
Collapse
|
15
|
Beesetti S. Ubiquitin Ligases in Control: Regulating NLRP3 Inflammasome Activation. FRONT BIOSCI-LANDMRK 2025; 30:25970. [PMID: 40152367 DOI: 10.31083/fbl25970] [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: 08/02/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 03/29/2025]
Abstract
Ubiquitin ligases play pivotal roles in the regulation of NLR family pyrin domain containing 3 (NLRP3) inflammasome activation, a critical process in innate immunity and inflammatory responses. This review explores the intricate mechanisms by which various E3 ubiquitin ligases exert both positive and negative influences on NLRP3 inflammasome activity through diverse post-translational modifications. Negative regulation of NLRP3 inflammasome assembly is mediated by several E3 ligases, including F-box and leucine-rich repeat protein 2 (FBXL2), tripartite motif-containing protein 31 (TRIM31), and Casitas B-lineage lymphoma b (Cbl-b), which induce K48-linked ubiquitination of NLRP3, targeting it for proteasomal degradation. Membrane-associated RING-CH 7 (MARCH7) similarly promotes K48-linked ubiquitination leading to autophagic degradation, while RING finger protein (RNF125) induces K63-linked ubiquitination to modulate NLRP3 function. Ariadne homolog 2 (ARIH2) targets the nucleotide-binding domain (NBD) domain of NLRP3, inhibiting its activation, and tripartite motif-containing protein (TRIM65) employs dual K48 and K63-linked ubiquitination to suppress inflammasome assembly. Conversely, Pellino2 exemplifies a positive regulator, promoting NLRP3 inflammasome activation through K63-linked ubiquitination. Additionally, ubiquitin ligases influence other components critical for inflammasome function. TNF receptor-associated factor 3 (TRAF3) mediates K63 polyubiquitination of apoptosis-associated speck-like protein containing a CARD (ASC), facilitating its degradation, while E3 ligases regulate caspase-1 activation and DEAH-box helicase 33 (DHX33)-NLRP3 complex formation through specific ubiquitination events. Beyond direct inflammasome regulation, ubiquitin ligases impact broader innate immune signaling pathways, modulating pattern-recognition receptor responses and dendritic cell maturation. Furthermore, they intricately control NOD1/NOD2 signaling through K63-linked polyubiquitination of receptor-interacting protein 2 (RIP2), crucial for nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) activation. Furthermore, we explore how various pathogens, including bacteria, viruses, and parasites, have evolved sophisticated strategies to hijack the host ubiquitination machinery, manipulating NLRP3 inflammasome activation to evade immune responses. This comprehensive analysis provides insights into the molecular mechanisms underlying inflammasome regulation and their implications for inflammatory diseases, offering potential avenues for therapeutic interventions targeting the NLRP3 inflammasome. In conclusion, ubiquitin ligases emerge as key regulators of NLRP3 inflammasome activation, exhibiting a complex array of functions that finely tune immune responses. Understanding these regulatory mechanisms not only sheds light on fundamental aspects of inflammation but also offers potential therapeutic avenues for inflammatory disorders and infectious diseases.
Collapse
Affiliation(s)
- Swarna Beesetti
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
16
|
Li J, Zhu C, Meng Y, Zhang L, Liu C, Qin Y, Chen M. Zika virus inhibits cell death by inhibiting the expression of NLRP3 and A20. J Virol 2025; 99:e0198024. [PMID: 39976465 PMCID: PMC11915814 DOI: 10.1128/jvi.01980-24] [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: 11/07/2024] [Accepted: 01/20/2025] [Indexed: 03/19/2025] Open
Abstract
Zika virus (ZIKV) is associated with microcephaly in neonates and neurological disorders in adults. Chronic ZIKV infection has been identified in the testes, indicating that the virus can lead to prolonged illness, yet its pathogenesis remains poorly understood. Here, we found that ZIKV infection does not induce significant cell death in mouse macrophages despite the critical role that cell death plays in the antiviral immune response. Furthermore, we discovered that ZIKV infection impairs the activation of the NLPR3-dependent inflammasome and inhibits apoptosis. Consequently, we investigated the regulatory mechanism of the NLRP3 inflammasome and apoptosis in the context of ZIKV infection. Our results revealed significant reductions in the protein expression levels of NLRP3 and A20, attributable to post-transcriptional or translational effects during ZIKV infection. These findings suggest that ZIKV infection may disrupt cell death pathways, leading to its pathogenicity.IMPORTANCEZika virus (ZIKV), first isolated from a nonhuman primate in Africa in 1947, was relatively understudied until 2016. By then, ZIKV had already been reported in more than 20 countries and territories. The infection poses a significant risk, as it is associated with microcephaly in infants and neurological disorders in adults; however, the underlying mechanisms responsible for these severe outcomes remain unclear. In this study, we demonstrate that ZIKV infection significantly reduces the expression of NLRP3 and A20 proteins through post-transcriptional or translational processes, which leads to inhibited cell death. These findings are critical because cell death plays a vital role in the host's antiviral immune response. Our findings highlight how ZIKV infection compromises essential cell death pathways, raising serious concerns about its pathogenesis. A comprehensive understanding of this disruption is vital for developing targeted interventions to mitigate the virus' impact on public health.
Collapse
Affiliation(s)
- Jian Li
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Changyang Zhu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yang Meng
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Linliang Zhang
- College of Life Sciences, Hubei University, Wuhan, China
| | - Cong Liu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yali Qin
- College of Life Sciences, Hubei University, Wuhan, China
| | - Mingzhou Chen
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
- College of Life Sciences, Hubei University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
| |
Collapse
|
17
|
Mandle HB, Jenab M, Gunter MJ, Tjønneland A, Olsen A, Dahm CC, Zhang J, Sugier PE, Rothwell J, Severi G, Kaaks R, Katzke VA, Schulze MB, Masala G, Sieri S, Panico S, Sacerdote C, Bonet C, Sánchez MJ, Amiano P, Huerta JM, Guevara M, Palmqvist R, Löwenmark T, Perez-Cornago A, Weiderpass E, Heath AK, Cross AJ, Vineis P, Hughes DJ, Fedirko V. Inflammation and gut barrier function-related genes and colorectal cancer risk in western European populations. Mutagenesis 2025; 40:48-60. [PMID: 38441165 PMCID: PMC11911009 DOI: 10.1093/mutage/geae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 04/04/2024] [Indexed: 03/21/2024] Open
Abstract
Gut barrier dysfunction and related inflammation are known to be associated with the development and progression of colorectal cancer (CRC). We investigated associations of 292 single-nucleotide polymorphisms (SNPs) from 27 genes related to endotoxins/lipopolysaccharide (LPS) sensing and tolerance, mucin synthesis, inflammation, and Crohn's disease with colon and rectal cancer risks. Incident CRC cases (N = 1374; colon = 871, rectum = 503) were matched 1:1 to controls nested within the European Prospective Investigation into Cancer and Nutrition cohort. Previously measured serum concentrations of gut barrier function and inflammation biomarkers (flagellin/LPS-specific immunoglobulins and C-reactive protein [CRP]) were available for a sub-set of participants (Ncases = 1001; Ncontrols = 667). Forty-two unique SNPs from 19 different genes were associated with serum biomarkers at Punadjusted ≤ 0.05 among controls. Among SNPs associated with a gut permeability score, 24 SNPs were in genes related to LPS sensing and mucin synthesis. Nine out of 12 SNPs associated with CRP were in genes related to inflammation or Crohn's disease. TLR4 was associated with colon cancer at the SNP level (nine SNPs, all Punadjusted ≤ 0.04) and at the gene level (Punadjusted ≤ 0.01). TLR4 rs10759934 was associated with rectal cancer but not colon cancer. Similarly, IL10 was associated with rectal cancer risk at an SNP and gene level (both Punadjusted ≤ 0.01), but not colon cancer. Genes and SNPs were selected a priori; therefore, we present unadjusted P-values. However, no association was statistically significant after multiple testing correction. This large and comprehensive study has identified gut barrier function and inflammation-related genes possibly contributing to CRC risk in European populations and is consistent with potential etiological links between host genetic background, gut barrier permeability, microbial endotoxemia, and CRC development.
Collapse
Affiliation(s)
- Hannah B Mandle
- Department of Epidemiology, Emory Rollins School of Public Health, Atlanta, GA 30322, USA
| | - Mazda Jenab
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France
| | - Marc J Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, SW7 2AZ, UK
| | - Anne Tjønneland
- Diet, Cancer and Health, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, 1353 Copenhagen K, Denmark
| | - Anja Olsen
- Department of Public Health, University of Copenhagen, 1353 Copenhagen K, Denmark
- Department of Public Health, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Christina C Dahm
- Department of Public Health, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Jie Zhang
- Department of Public Health, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Pierre-Emmanuel Sugier
- Université Paris-Saclay, UVSQ, Inserm ‘Exposome and Heredity’ team, CESP U1018, 94807 Villejuif Cedex, France
- Laboratoire de Mathématiques et de leurs Applications de Pau E2S UPPA, CNRS, 64013 Pau Cedex, France
| | - Joseph Rothwell
- Université Paris-Saclay, UVSQ, Inserm ‘Exposome and Heredity’ team, CESP U1018, 94807 Villejuif Cedex, France
| | - Gianluca Severi
- Université Paris-Saclay, UVSQ, Inserm ‘Exposome and Heredity’ team, CESP U1018, 94807 Villejuif Cedex, France
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
| | - Verena A Katzke
- Division of Cancer Epidemiology, German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition, Potsdam-Rehbruecke, 14469 Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, 14469 Nuthetal, Germany
| | - Giovanna Masala
- Institute for Cancer Research, Prevention and Clinical Network (ISPRO), 50139 Florence, Italy
| | - Sabina Sieri
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy
| | - Salvatore Panico
- Dipartimento Di Medicina Clinica E Chirurgia, Federico II University, 80131 Naples, Italy
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, AOU Città della Salute e della Scienza University Hospital, 10126 Turin, Italy
| | - Catalina Bonet
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO), L’Hospitalet de Llobregat, 0890x Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 0890x Barcelona, Spain
| | - Maria-Jose Sánchez
- Escuela Andaluza de Salud Pública (EASP), 18011 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.18011 Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Granada, 18071 Granada, Spain
| | - Pilar Amiano
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Ministry of Health of the Basque Government, Sub Directorate for Public Health and Addictions of Gipuzkoa, BioGipuzkoa Health Research Institute, Epidemiology of Chronic and Communicable Diseases Group, 20014 Donostia – San Sebastian, Spain
| | - José María Huerta
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Department of Epidemiology, Murcia Regional Health Council-IMIB, 30120, El Palmar, Murcia, Spain
| | - Marcela Guevara
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Instituto de Salud Pública y Laboral de Navarra, 31003 Pamplona, Navarra, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Navarra, Spain
| | - Richard Palmqvist
- Department of Medical Biosciences, Umea University, 901 87 Umeå, Sweden
| | - Thyra Löwenmark
- Department of Medical Biosciences, Umea University, 901 87 Umeå, Sweden
| | - Aurora Perez-Cornago
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Elisabete Weiderpass
- Office of the Director, International Agency for Research on Cancer, 69366 Lyon Cedex 07, France
| | - Alicia K Heath
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, SW7 2AZ, UK
| | - Amanda J Cross
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, SW7 2AZ, UK
| | - Paolo Vineis
- MRC Centre for Environment and Health, School of public Health, Imperial College London, London W2 1PG, UK
- Italian Institute for Genomic Medicine (IIGM), 10060 Candiolo TO,Italy
| | - David J Hughes
- Cancer Biology and Therapeutics Group, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Veronika Fedirko
- Department of Epidemiology, Emory Rollins School of Public Health, Atlanta, GA 30322, USA
- Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, 77030 Houston, TX, USA
| |
Collapse
|
18
|
Cui B, Ai L, Lei M, Duan Y, Tang C, Zhang J, Gao Y, Li X, Zhu C, Zhang Y, Zhu X, Isobe T, Yang W, Göttgens B, Zhu P. Single-cell epigenetic and clonal analysis decodes disease progression in pediatric acute myeloid leukemia. Blood 2025; 145:1211-1224. [PMID: 39661948 PMCID: PMC11923433 DOI: 10.1182/blood.2024025618] [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: 06/03/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024] Open
Abstract
ABSTRACT Pediatric acute myeloid leukemia (pAML) is a clonal disease with recurrent genetic alterations that affect epigenetic states. However, the implications of epigenetic dysregulation in disease progression remain unclear. Here, we interrogated single-cell and clonal level chromatin accessibility of bone marrow samples from 28 patients with pAML representing multiple subtypes using mitochondrial single-cell assay for transposase-accessible chromatin with sequencing, which revealed distinct differentiation hierarchies and abnormal chromatin accessibility in a subtype-specific manner. Innate immune signaling was commonly enhanced across subtypes and related to improved advantage of clonal competition and unfavorable prognosis, with further reinforcement in a relapse-associated leukemia stem cell-like population. We identified a panel of 31 innate immunity-related genes to improve the risk classification of patients with pAML. By comparing paired diagnosis and postchemotherapy relapse samples, we showed that primitive cells significantly reduced major histocompatibility complex class II signaling, suggesting an immune evasion mechanism to facilitate their expansion at relapse. Key regulators orchestrating cell cycle dysregulation were identified to contribute to pAML relapse in drug-resistant clones. Our work establishes the single-cell chromatin accessibility landscape at clonal resolution and reveals the critical involvement of epigenetic disruption, offering insights into classification and targeted therapies of patients with pAML.
Collapse
Affiliation(s)
- Boyu Cui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Lanlan Ai
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Minghui Lei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yongjuan Duan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Chao Tang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jingliao Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yan Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xuan Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Caiying Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yingchi Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xiaofan Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Tomoya Isobe
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Department of Haematology, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
| | - Wenyu Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Berthold Göttgens
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Department of Haematology, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
| | - Ping Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
19
|
Mahjoubin-Tehran M, Rezaei S, Butler AE, Sahebkar A. Decoy oligonucleotides targeting NF-κB: a promising therapeutic approach for inflammatory diseases. Inflamm Res 2025; 74:47. [PMID: 40047902 DOI: 10.1007/s00011-025-02021-8] [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: 08/08/2024] [Revised: 02/11/2025] [Accepted: 03/02/2025] [Indexed: 05/13/2025] Open
Abstract
Nuclear factor-kappa B (NF-κB) transcription factor plays a crucial function in controlling several cellular processes, including the production of inflammatory mediators. The aberrant activation of this transcription factor and its signaling pathway is associated with the pathophysiology of many diseases. Therefore, discovering drugs that target NF-κB is crucial for treating various diseases. Decoy oligonucleotides (decoy ONs) are a pharmacological approach that specifically inhibits NF-κB activation and are used to treat several inflammatory diseases. Decoys that target NF-κB have been shown to enhance radiosensitivity and drug sensitivity in vitro and strongly block IL-6 and IL-8 gene expression induced by TNF-α in experimental cell systems. In vivo, NF-κB decoy reduced atherosclerotic plaque, prevented atopic dermatitis and extended cardiac transplant survival. Decoys have the potential to be used in clinical applications, but they face several challenges. To overcome these limitations, researchers have conducted studies on chemical modifications and delivery techniques. Innovative compounds that target NF-κB, such as NF-κB-decoy-based sensor-containing models, phosphorothioate hairpin-modified oligonucleotides, and peptide nucleic acid (PNA)-based transcription factor decoys, are very attractive. This research aims to explore the use of decoys to combat NF-κB in various disorders.
Collapse
Affiliation(s)
| | - Samaneh Rezaei
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland Bahrain, Adliya, 15503, Bahrain
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai, India.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
20
|
Ma ZF, Fu C, Lee YY. The Modulatory Role of Bioactive Compounds in Functional Foods on Inflammation and Metabolic Pathways in Chronic Diseases. Foods 2025; 14:821. [PMID: 40077524 PMCID: PMC11899172 DOI: 10.3390/foods14050821] [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: 02/01/2025] [Revised: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Chronic diseases are major contributors to global morbidity and mortality. More than 70% of deaths worldwide are caused by chronic diseases, including cardiovascular diseases (CVDs), obesity, type 2 diabetes, and cancer. These diseases are characterised by chronic low-grade inflammation and metabolic dysregulation. Incorporating functional foods into daily diet has been suggested as a complementary strategy to promote health and lower the risk of non-communicable diseases. Functional foods, known as foods that confer health benefits beyond basic nutrition, have been reported to exhibit preventive and therapeutic benefits such as anti-inflammatory properties for human health. Therefore, the aim of this state-of-the-art review will synthesise the findings from recent and high-quality studies that investigated the modulatory role of some commonly reported bioactive active compounds, such as polyphenols, omega-3 fatty acids, probiotics, and prebiotics, in inflammation and metabolic pathways.
Collapse
Affiliation(s)
- Zheng Feei Ma
- Centre for Public Health and Wellbeing, School of Health and Social Wellbeing, College of Health, Science and Society, University of the West of England, Bristol BS16 1QY, UK
| | - Caili Fu
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Yeong Yeh Lee
- School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 15200, Malaysia
| |
Collapse
|
21
|
Mammoliti O, Carbajo R, Perez-Benito L, Yu X, Prieri MLC, Bontempi L, Embrechts S, Paesmans I, Bassi M, Bhattacharya A, Cañellas S, De Hoog S, Demin S, Gijsen HJM, Hache G, Jacobs T, Jerhaoui S, Leenaerts J, Lutter FH, Mahieu M, Matico R, Miller R, Oehlrich D, Perrier M, Ryabchuk P, Schepens W, Sharma S, Somers M, Suarez J, Surkyn M, Van Opdenbosch N, Verhulst T, Bottelbergs A. Discovery of Potent and Brain-Penetrant Bicyclic NLRP3 Inhibitors with Peripheral and Central In Vivo Activity. J Med Chem 2025; 68:4848-4887. [PMID: 39932543 DOI: 10.1021/acs.jmedchem.4c03108] [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: 02/28/2025]
Abstract
NLRP3 is a danger sensor protein responsible for inflammasome activation. This leads to pro-inflammatory cytokines release, like IL-1β, and pyroptosis, a regulated cell death. Mounting evidence associates excessive NLRP3 activation to neurodegenerative conditions, such as Alzheimer's and Parkinson's diseases. Thus, NLRP3 inhibitors could potentially provide therapeutic benefit for these disorders. We describe here the evolution of inhibitors relying on a pyridazine-based motif for their key interactions with NLRP3. A Cryo-EM structure helped optimizing protein-ligand complementarity. Subsequently, conformational NMR studies pointed the efforts toward 5,6-bicyclic cores that allowed a balance between brain penetration and undesirable properties, such as hERG inhibition. The effort culminated in compound 19, which showed moderate (mouse) to good (rat) brain penetration and was active at low dose in an LPS challenge model. Importantly, an earlier compound was active in a central neuroinflammation model providing a valuable proof of concept for NLRP3 inhibition.
Collapse
Affiliation(s)
- Oscar Mammoliti
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Rodrigo Carbajo
- Janssen-Cilag S.A., A Johnson & Johnson Company, C/Jarama 75A, 45007 Toledo, Spain
| | - Laura Perez-Benito
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Xiaodi Yu
- Janssen Research & Development, LLC, A Johnson & Johnson Company, Spring House, Pennsylvania 19477, United States
| | - Marion L C Prieri
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Leonardo Bontempi
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Sofie Embrechts
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Ine Paesmans
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Michela Bassi
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Anindya Bhattacharya
- Janssen Research & Development, LLC, A Johnson & Johnson Company, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Santiago Cañellas
- Janssen-Cilag S.A., A Johnson & Johnson Company, C/Jarama 75A, 45007 Toledo, Spain
| | - Saskia De Hoog
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Samuël Demin
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Harrie J M Gijsen
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Geerwin Hache
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Tom Jacobs
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Soufyan Jerhaoui
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Joseph Leenaerts
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Ferdinand H Lutter
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Michel Mahieu
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Rosalie Matico
- Janssen Research & Development, LLC, A Johnson & Johnson Company, Spring House, Pennsylvania 19477, United States
| | - Robyn Miller
- Janssen Research & Development, LLC, A Johnson & Johnson Company, Spring House, Pennsylvania 19477, United States
| | - Daniel Oehlrich
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Mathieu Perrier
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Pavel Ryabchuk
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Wim Schepens
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Sujata Sharma
- Janssen Research & Development, LLC, A Johnson & Johnson Company, Spring House, Pennsylvania 19477, United States
| | - Marijke Somers
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Javier Suarez
- Janssen Research & Development, LLC, A Johnson & Johnson Company, Spring House, Pennsylvania 19477, United States
| | - Michel Surkyn
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | | | - Tinne Verhulst
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Astrid Bottelbergs
- Janssen Pharmaceutica NV, A Johnson & Johnson Company, Turnhoutseweg 30, Beerse 2340, Belgium
| |
Collapse
|
22
|
Bruinjé TC, LeBlanc SJ. Invited Review: Inflammation and Health in the Transition Period Influence Reproductive Function in Dairy Cows. Animals (Basel) 2025; 15:633. [PMID: 40075916 PMCID: PMC11898178 DOI: 10.3390/ani15050633] [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: 12/30/2024] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
In the early postpartum period, dairy cows undergo significant adaptations in Ca and lipid metabolism, immune function, and inflammatory processes. Concurrent exposure to endotoxins from the uterus, gastrointestinal tract, or mammary gland increases the risk of disease and reproductive problems. Metabolic and inflammatory imbalances during this phase can have both immediate and long-term effects on reproductive health. Associations between metabolic disorders and reproductive outcomes are often confounded by immune activation and systemic inflammation. However, optimal markers, thresholds, and durations for identifying maladaptation and predicting adverse health or reproductive outcomes remain unclear. This narrative review examines key physiological changes during the transition period, including hypocalcemia, lipid mobilization, immune activation, systemic inflammation, and uterine disease. We discuss how these events may affect the dominant follicle, corpus luteum, oocyte, and uterus, potentially leading to prolonged anovulation, reduced estrus expression, impaired response to synchronization protocols, lower progesterone concentrations, and compromised fertility. Understanding these mechanisms will support the development of strategies to monitor, prevent, and mitigate the impacts of transition-related maladaptation on reproductive performance. Such advancements can enhance the health and fertility of high-producing dairy cows.
Collapse
Affiliation(s)
- Tony C. Bruinjé
- Department of Dairy and Food Science, South Dakota State University, Brookings, SD 57007, USA
- Department of Population Medicine, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Stephen J. LeBlanc
- Department of Population Medicine, University of Guelph, Guelph, ON N1G 2W1, Canada;
| |
Collapse
|
23
|
Brisse ME, Hickman HD. Viral Infection and Dissemination Through the Lymphatic System. Microorganisms 2025; 13:443. [PMID: 40005808 PMCID: PMC11858409 DOI: 10.3390/microorganisms13020443] [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: 01/23/2025] [Revised: 02/11/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025] Open
Abstract
Many viruses induce viremia (virus in the blood) and disseminate throughout the body via the bloodstream from the initial infection site. However, viruses must often pass through the lymphatic system to reach the blood. The lymphatic system comprises a network of vessels distinct from blood vessels, along with interconnected lymph nodes (LNs). The complex network has become increasingly appreciated as a crucial host factor that contributes to both the spread and control of viral infections. Viruses can enter the lymphatics as free virions or along with migratory cells. Once virions arrive in the LN, sinus-resident macrophages remove infectious virus from the lymph. Depending on the virus, macrophages can eliminate infection or propagate the virus. A virus released from an LN is eventually deposited into the blood. This unique pathway highlights LNs as targets for viral infection control and for modulation of antiviral response development. Here, we review the lymphatic system and viruses that disseminate through this network. We discuss infection of the LN, the generation of adaptive antiviral immunity, and current knowledge of protection within the infected node. We conclude by sharing insights from ongoing efforts to optimize lymphatic targeting by vaccines and pharmaceuticals. Understanding the lymphatic system's role during viral infection enhances our knowledge of antiviral immunity and virus-host interactions and reveals potential targets for next-generation therapies.
Collapse
Affiliation(s)
| | - Heather D. Hickman
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA;
| |
Collapse
|
24
|
Wu CJ. NEMO Family of Proteins as Polyubiquitin Receptors: Illustrating Non-Degradative Polyubiquitination's Roles in Health and Disease. Cells 2025; 14:304. [PMID: 39996775 PMCID: PMC11854354 DOI: 10.3390/cells14040304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/26/2025] Open
Abstract
The IκB kinase (IKK) complex plays a central role in many signaling pathways that activate NF-κB, which turns on a battery of genes important for immune response, inflammation, and cancer development. Ubiquitination is one of the most prevalent post-translational modifications of proteins and is best known for targeting substrates for proteasomal degradation. The investigations of NF-κB signaling pathway primed the unveiling of the non-degradative roles of protein ubiquitination. The NF-κB-essential modulator (NEMO) is the IKK regulatory subunit that is essential for IKK activation by diverse intrinsic and extrinsic stimuli. The studies centered on NEMO as a polyubiquitin-binding protein have remarkably advanced understandings of how NEMO transmits signals to NF-κB activation and have laid a foundation for determining the molecular events demonstrating non-degradative ubiquitination as a major driving element in IKK activation. Furthermore, these studies have largely solved the enigma that IKK can be activated by diverse pathways that employ distinct sets of intermediaries in transmitting signals. NEMO and NEMO-related proteins that include optineurin, ABIN1, ABIN2, ABIN3, and CEP55, as non-degradative ubiquitin chain receptors, play a key role in sensing and transmitting ubiquitin signals embodied in different topologies of polyubiquitin chains for a variety of cellular processes and body responses. Studies of these multifaceted proteins in ubiquitin sensing have promoted understanding about the functions of non-degradative ubiquitination in intracellular signaling, protein trafficking, proteostasis, immune response, DNA damage response, and cell cycle control. In this review, I will also discuss how dysfunction in the NEMO family of protein-mediated non-degradative ubiquitin signaling is associated with various diseases, including immune disorders, neurodegenerative diseases, and cancer, and how microbial virulence factors target NEMO to induce pathogenesis or manipulate host response. A profound understanding of the molecular bases for non-degradative ubiquitin signaling will be valuable for developing tailored approaches for therapeutic purposes.
Collapse
Affiliation(s)
- Chuan-Jin Wu
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
25
|
Otálora-Alcaraz A, Reilly T, Oró-Nolla M, Sun MC, Costelloe L, Kearney H, Patra PH, Downer EJ. The NLRP3 inflammasome: A central player in multiple sclerosis. Biochem Pharmacol 2025; 232:116667. [PMID: 39647604 DOI: 10.1016/j.bcp.2024.116667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/06/2024] [Accepted: 11/25/2024] [Indexed: 12/10/2024]
Abstract
Multiple sclerosis (MS) is a neurological autoimmune condition associated with many symptoms including spasticity, pain, limb numbness and weakness. It is characterised by inflammatory demyelination and axonal degeneration of the brain and spinal cord. A range of disease-modifying therapies (DMTs) are available to suppress inflammatory disease activity in MS, however, there is a pressing need for new therapeutic avenues as DMTs have a limited ability to suppress confirmed disability progression. A body of literature indicates that innate immune inflammation is linked to MS progression. The nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain containing protein 3 (NLRP3) inflammasome has a well-established function in innate immunity which is closely associated with the pathogenesis of neuroinflammatory conditions. Evidence suggests that the inflammasome may be a therapeutic target in disorders such as MS and at present, inhibitors of the NLRP3 inflammasome are in pre-clinical development. Therefore, this review systematically highlights the pathogenic role of inflammasomes in MS, presenting an overview of research evidence linking inflammasome-related polymorphisms to MS susceptibility, and gathering evidence investigating NLRP3 biomarkers in MS. The role of the NLRP3 inflammasome in murine models of MS is furthermore discussed. Finally, a significant component of this review focuses on evidence that NLRP3 signalling components are novel drug targets in MS. Overall this review defines the role of the inflammasome in MS pathogenesis and identifies inflammasome inhibitor targets that warrant full investigation in MS and related disorders.
Collapse
Affiliation(s)
- Almudena Otálora-Alcaraz
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Thomas Reilly
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Martí Oró-Nolla
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Melody Cui Sun
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Lisa Costelloe
- Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Hugh Kearney
- MS Unit, Department of Neurology, St. James's Hospital, Dublin, Ireland; Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - Pabitra H Patra
- Transpharmation Ltd., London Biosciences Innovation Centre, London, United Kingdom
| | - Eric J Downer
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
26
|
López-Hernández R, de la Torre-Álamo MM, García-Bueno B, Baroja-Mazo A, Fenoy FJ, Cuevas S. Inflammasomes in Alzheimer's Progression: Nrf2 as a Preventive Target. Antioxidants (Basel) 2025; 14:121. [PMID: 40002308 PMCID: PMC11851705 DOI: 10.3390/antiox14020121] [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: 12/19/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
Current knowledge about Alzheimer's disease highlights the accumulation of β-amyloid plaques (Aβ1-42) and neurofibrillary tangles composed of hyperphosphorylated Tau, which lead to the loss of neuronal connections. Microglial activation and the release of inflammatory mediators play a significant role in the progression of Alzheimer's pathology. Recent advances have identified the involvement of inflammasomes, particularly NOD-like receptor NLR family pyrin domain containing 3 (NLRP3), whose activation promotes the release of proinflammatory cytokines and triggers pyroptosis, exacerbating neuroinflammation. Aggregates of Aβ1-42 and hyperphosphorylated Tau have been shown to activate these inflammasomes, while the apoptosis-associated speck-like protein (ASC) components form aggregates that further accelerate Aβ aggregation. Defects in the autophagic clearance of inflammasomes have also been implicated in Alzheimer's disease, contributing to sustained inflammation. This review explores strategies to counteract inflammation in Alzheimer's, emphasizing the degradation of ASC specks and the inhibition of NLRP3 inflammasome activation. Notably, the nuclear factor erythroid 2-related factor 2 (Nrf2) transcription factor emerges as a promising therapeutic target due to its dual role in mitigating oxidative stress and directly inhibiting NLRP3 inflammasome formation. By reducing inflammasome-driven inflammation, Nrf2 offers significant potential for addressing the neuroinflammatory aspects of Alzheimer's disease.
Collapse
Affiliation(s)
- Rubén López-Hernández
- Molecular Inflammation Group, Pathophysiology of the Inflammation and Oxidative Stress Lab, Biomedical Research Institute of Murcia (IMIB), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain;
| | - María Magdalena de la Torre-Álamo
- Molecular Inflammation Group, Digestive and Endocrine Surgery and Transplantation of Abdominal Organs, Biomedical Research Institute of Murcia (IMIB), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain; (M.M.d.l.T.-Á.); (B.G.-B.); (A.B.-M.)
| | - Belén García-Bueno
- Molecular Inflammation Group, Digestive and Endocrine Surgery and Transplantation of Abdominal Organs, Biomedical Research Institute of Murcia (IMIB), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain; (M.M.d.l.T.-Á.); (B.G.-B.); (A.B.-M.)
| | - Alberto Baroja-Mazo
- Molecular Inflammation Group, Digestive and Endocrine Surgery and Transplantation of Abdominal Organs, Biomedical Research Institute of Murcia (IMIB), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain; (M.M.d.l.T.-Á.); (B.G.-B.); (A.B.-M.)
| | - Francisco Jose Fenoy
- Department of Physiology, Faculty of Medicine, University of Murcia, 30120 Murcia, Spain;
| | - Santiago Cuevas
- Molecular Inflammation Group, Pathophysiology of the Inflammation and Oxidative Stress Lab, Biomedical Research Institute of Murcia (IMIB), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain;
| |
Collapse
|
27
|
Atre R, Obukhov AG, Majmudar CY, Nair K, White FA, Sharma R, Siddiqi F, Faisal SM, Varma VP, Hassan MI, Mohammad T, Darwhekar GN, Baig MS. Dorzolamide intermediates with potential anti-inflammatory activity. Eur J Pharmacol 2025; 987:177160. [PMID: 39631651 DOI: 10.1016/j.ejphar.2024.177160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Dorzolamide (DZD), a Carbonic anhydrase (CA) inhibitor clinically used to lower intraocular pressure, exhibits anti-inflammatory effects owing to the drug's ability to inhibit the TIR domain-containing adaptor protein (TIRAP)-mediated signalling in macrophages. Here, we investigated whether DZD intermediates also demonstrate any anti-inflammatory property like DZD but with a reduced inhibition of CA. We found that several intermediates of DZD show increased binding to TIRAP at the common interface of kinases, such as Protein kinase C-delta (PKCδ) and Bruton's tyrosine kinase (BTK). Such binding results in a decreased activity of TIRAP, p38 Mitogen-activating protein kinases (MAPK), and p65, which are essential for major inflammatory signaling pathways. Remarkably, the DZD intermediates were more effective than DZD in decreasing the mRNA expression levels of pro-inflammatory cytokines in Lipopolysaccharide (LPS)-stimulated RAW 264.7 cells. The DZD intermediates also exhibit a reduced binding energy to CA II and CA IV, highlighting their improved specificity as anti-inflammatory compounds with decreased unwanted biological effects. Furthermore, we validated the anti-inflammatory effect of the most efficient DZD intermediate, DRZ V, in a model of mouse sepsis. DRZ V-treated septic mice exhibited improved survival compared to DZD-treated septic mice. Our data indicate that the tested DZD intermediates are more effectual in dampening TIRAP-mediated inflammatory signaling as compared to DZD. Thus, DZD intermediates may be a promising option for developing novel anti-inflammatory therapeutics.
Collapse
Affiliation(s)
- Rajat Atre
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Alexander G Obukhov
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - Fletcher A White
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rahul Sharma
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Faaiza Siddiqi
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Syed M Faisal
- Laboratory of Vaccine Immunology, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
| | - Vivek P Varma
- Laboratory of Vaccine Immunology, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Gajanan N Darwhekar
- Acropolis Institute of Pharmaceutical Education and Research (AIPER), Indore, MP, 453771, India
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India.
| |
Collapse
|
28
|
Kuramochi M, Karim MR, Izawa T, Kuwamura M, Yamate J. High mobility group box1 as a danger signal inducing the infiltration of neutrophils and macrophages in thioacetamide-induced rat liver injury. J Toxicol Pathol 2025; 38:49-58. [PMID: 39839722 PMCID: PMC11745500 DOI: 10.1293/tox.2024-0055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/28/2024] [Indexed: 01/23/2025] Open
Abstract
The liver, a major organ involved in substance metabolism, is highly susceptible to toxicity induced by chemicals and their metabolites. Although damage-associated molecular patterns (DAMPs) have been implicated in the development of sterile inflammation following cell injury, their involvement in chemically induced hepatocellular injury remains underexplored. This study aimed to determine the role of high-mobility group box 1 (HMGB1), a DAMP, in a rat model of liver injury treated with thioacetamide, a hepatotoxicant. The rats were administered thioacetamide and treated with HMGB1 neutralizing antibody. Histopathological analysis revealed the absence of significant differences between control rats and HMGB1 neutralizing antibody-treated rats. However, HMGB1 neutralizing antibody-treated rats showed a reduction in the hepatic devitalization enzymes, a decrease in the number of anti-inflammatory cluster of differentiation CD163+ M2 macrophages and neutrophils in the injured area, and a decrease in cytokine expression. These results suggest that HMGB1 leads to the progression of inflammation after chemically induced hepatocyte injury and may represent a therapeutic target for mitigating such injury.
Collapse
Affiliation(s)
- Mizuki Kuramochi
- Laboratory of Veterinary Pathology, Osaka Metropolitan
University, 1-58 Rinku-Ourai-Kita, Izumisano City, Osaka 598-8531, Japan
| | - Mohammad Rabiul Karim
- Laboratory of Veterinary Pathology, Osaka Metropolitan
University, 1-58 Rinku-Ourai-Kita, Izumisano City, Osaka 598-8531, Japan
- Laboratory of Distorted Anatomy, Department of Anatomy and
Histology, Faculty of Veterinary Science, Bangladesh Agricultural University,
Mymensingh-2202, Bangladesh
| | - Takeshi Izawa
- Laboratory of Veterinary Pathology, Osaka Metropolitan
University, 1-58 Rinku-Ourai-Kita, Izumisano City, Osaka 598-8531, Japan
| | - Mitsuru Kuwamura
- Laboratory of Veterinary Pathology, Osaka Metropolitan
University, 1-58 Rinku-Ourai-Kita, Izumisano City, Osaka 598-8531, Japan
| | - Jyoji Yamate
- Laboratory of Veterinary Pathology, Osaka Metropolitan
University, 1-58 Rinku-Ourai-Kita, Izumisano City, Osaka 598-8531, Japan
| |
Collapse
|
29
|
Zheng B, Chen J, Xu Y, Wu W, Zhu Y, Cai W, Lin W, Shi C. Poly (β-amino esters)/Mobil Composition of Matter 41-mediated delivery of siIL-1β alleviates deep vein thrombosis in rat hind limbs. J Biomater Appl 2025; 39:648-660. [PMID: 39213651 DOI: 10.1177/08853282241280376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Introduction: Deep vein thrombosis (DVT) is a major cause of cardiovascular disease-related deaths worldwide and is considered a thrombotic inflammatory disorder. IL-1β, as a key promoter of venous thrombus inflammation, is a potential target for DVT treatment. Constructing a nanocarrier system for intracellular delivery of siIL-1β to silence IL-1β may be an effective strategy for alleviating DVT. Methods: ELISA was used to detect the expression levels of IL-1β and t-PA in the serum of DVT patients and healthy individuals. In vitro, HUVEC cells were treated with IL-1β, and changes in VWF and t-PA expression levels were assessed. PBAE/MCM-41@siIL-1β (PM@siIL-1β) nano-complexes were synthesized, the characterization and biocompatibility of PM@siIL-1β were evaluated. A rat hind limb DVT model was established, and PM@siIL-1β was used to treat DVT rats. Morphology of the inferior vena cava, endothelial cell count, IL-1β, vWF, and t-PA levels, as well as changes in the p38 MAPK and NF-κB pathways, were examined in the different groups. Results: IL-1β and t-PA were highly expressed in DVT patients, and IL-1β treatment induced a decrease in VWF levels and an increase in t-PA levels in HUVEC cells. The synthesized PM@siIL-1β exhibited spherical shape, good stability, high encapsulation efficiency, and high drug loading capacity, with excellent biocompatibility. In the DVT model rats, the inferior vena cava was filled with blood clots, endothelial cells increased, IL-1β and VWF levels significantly increased, while t-PA levels were significantly downregulated. Treatment with PM@siIL-1β resulted in reduced thrombus formation, decreased endothelial cell count, and reversal of IL-1β, VWF, and t-PA levels. Furthermore, PM@siIL-1β treatment significantly inhibited p38 phosphorylation and upregulation of NF-κB expression in the DVT model group. Conclusion: IL-1β can be considered a therapeutic target for suppressing DVT inflammation. The synthesized PM@siIL-1β achieved efficient delivery and gene silencing of siIL-1β, demonstrating good therapeutic effects on rat hind limb DVT, including anti-thrombotic and anti-inflammatory effects, potentially mediated through the p38 MAPK and NF-κB pathways.
Collapse
Affiliation(s)
- Bingru Zheng
- Department of Interventional Vascular Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinjie Chen
- Department of Nephrology, Rui'an Third People's Hospital, Wenzhou, China
| | - Yizhou Xu
- Department of Interventional Vascular Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wanrui Wu
- Department of Interventional Vascular Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yu Zhu
- Department of Interventional Vascular Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei Cai
- Department of Interventional Vascular Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weili Lin
- Department of Ultrasound Imaging, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Changsheng Shi
- Department of Interventional Vascular Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
30
|
Ishino F, Itoh J, Matsuzawa A, Irie M, Suzuki T, Hiraoka Y, Yoshikawa M, Kaneko-Ishino T. RTL4, a Retrovirus-Derived Gene Implicated in Autism Spectrum Disorder, Is a Microglial Gene That Responds to Noradrenaline in the Postnatal Brain. Int J Mol Sci 2024; 25:13738. [PMID: 39769499 PMCID: PMC11678650 DOI: 10.3390/ijms252413738] [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: 10/23/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Retrotransposon Gag-like 4 (RTL4), a gene acquired from a retrovirus, is a causative gene in autism spectrum disorder. Its knockout mice exhibit increased impulsivity, impaired short-term spatial memory, failure to adapt to novel environments, and delayed noradrenaline (NA) recovery in the frontal cortex. However, due to its very low expression in the brain, it remains unknown which brain cells express RTL4 and its dynamics in relation to NA. We addressed these issues using knock-in mice carrying endogenous Rtl4 fused to Venus, which encodes a fluorescent protein. The RTL4-Venus fusion protein was detected as a secreted protein in the midbrain, hypothalamus, hippocampus and amygdala in the postnatal brain. Its signal intensity was high during critical periods of neonatal adaptation to novel environments. It was upregulated by various stimuli, including isoproterenol administration, whereas it was decreased by anesthesia but was maintained by milnacipran administration, suggesting its highly sensitive response to stressors, possible dependence on the arousal state and involvement in the NA reuptake process. In vitro mixed glial culture experiments demonstrated that Rtl4 is a microglial gene and suggested that RTL4 secretion responds rapidly to isoproterenol. Microglial RTL4 plays an important role in the NA response and possibly in the development of the NAergic neuronal network in the brain.
Collapse
Affiliation(s)
- Fumitoshi Ishino
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (A.M.); (M.I.)
| | - Johbu Itoh
- Department of Neurology, School of Medicine, Tokai University School of Medicine, Isehara 259-1193, Japan;
| | - Ayumi Matsuzawa
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (A.M.); (M.I.)
| | - Masahito Irie
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (A.M.); (M.I.)
- Faculty of Nursing, Tokai University School of Medicine, Isehara 259-1193, Japan
| | - Toru Suzuki
- Laboratory of Genome Editing for Biomedical Research, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (T.S.); (Y.H.)
| | - Yuichi Hiraoka
- Laboratory of Genome Editing for Biomedical Research, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; (T.S.); (Y.H.)
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| | - Masanobu Yoshikawa
- Department of Clinical Pharmacology, Tokai University School of Medicine, Isehara 259-1193, Japan;
| | - Tomoko Kaneko-Ishino
- Faculty of Nursing, Tokai University School of Medicine, Isehara 259-1193, Japan
| |
Collapse
|
31
|
Mackay A, Velcicky J, Gommermann N, Mattes H, Janser P, Wright M, Dubois C, Brenneisen S, Ilic S, Vangrevelinghe E, Stiefl N, Boettcher A, Schoenboerner M, Vogelsanger M, Muller-Bentz S, Kamke M, Rubert J, Kauffmann M, Desrayaud S, Trunzer M, Srinivas H, Hinniger A, von Burg N, Beltz K, Dekker C, Farady CJ. Discovery of NP3-253, a Potent Brain Penetrant Inhibitor of the NLRP3 Inflammasome. J Med Chem 2024; 67:20780-20798. [PMID: 39574318 DOI: 10.1021/acs.jmedchem.4c02350] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Activation of the NLRP3 inflammasome in response to danger signals is a key innate immune mechanism and results in the production of the pro-inflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18) as well as pyroptotic cell death. Aberrant NLRP3 activation has been linked to many acute and chronic conditions ranging from atherosclerosis to Alzheimer's disease and cancer, and based on the clinical success of IL-1-targeting therapies, NLRP3 has emerged as an attractive therapeutic target. Herein we describe our discovery, characterization, and structure-based optimization of a pyridazine-based series of NLRP3 inhibitors initiating from an high-throughput screening campaign. The scaffold, exemplified by lead molecule NP3-253, has excellent potency and physicochemical and pharmacokinetic properties, including good brain penetration. The establishment of pharmacokinetic/pharmacodynamic relationships in the periphery and central nervous system in mechanistic models facilitates the use of NP3-253 as a tool to further interrogate the biology of NLRP3 in peripheral and neuroinflammatory models.
Collapse
Affiliation(s)
- Angela Mackay
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Juraj Velcicky
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | | | - Henri Mattes
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Philipp Janser
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Michael Wright
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Celine Dubois
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | | | - Slavica Ilic
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | | | | | | | | | | | | | - Marion Kamke
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Joelle Rubert
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | | | | | - Markus Trunzer
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | | | | | | | - Karen Beltz
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Carien Dekker
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | | |
Collapse
|
32
|
Zhao Z, Ruan S, Li Y, Qi T, Qi Y, Huang Y, Liu Z, Ruan Q, Ma Y. The Influence of Extra-Ribosomal Functions of Eukaryotic Ribosomal Proteins on Viral Infection. Biomolecules 2024; 14:1565. [PMID: 39766272 PMCID: PMC11674327 DOI: 10.3390/biom14121565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/25/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
The eukaryotic ribosome is a large ribonucleoprotein complex consisting of four types of ribosomal RNA (rRNA) and approximately 80 ribosomal proteins (RPs), forming the 40S and 60S subunits. In all living cells, its primary function is to produce proteins by converting messenger RNA (mRNA) into polypeptides. In addition to their canonical role in protein synthesis, RPs are crucial in controlling vital cellular processes such as cell cycle progression, cellular proliferation, differentiation, DNA damage repair, genome structure maintenance, and the cellular stress response. Viruses, as obligate intracellular parasites, depend completely on the machinery of the host cell for their replication and survival. During viral infection, RPs have been demonstrated to perform a variety of extra-ribosomal activities, which are especially important in viral disease processes. These functions cover a wide range of activities, ranging from controlling inflammatory responses and antiviral immunity to promoting viral replication and increasing viral pathogenicity. Deciphering the regulatory mechanisms used by RPs in response to viral infections has greatly expanded our understanding of their functions outside of the ribosome. Furthermore, these findings highlight the promising role of RPs as targets for the advancement of antiviral therapies and the development of novel antiviral approaches. This review comprehensively examines the many functions of RPs outside of the ribosome during viral infections and provides a foundation for future research on the host-virus interaction.
Collapse
Affiliation(s)
- Zhongwei Zhao
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang 110004, China; (Z.Z.); (T.Q.); (Y.Q.); (Y.H.); (Z.L.)
| | - Shan Ruan
- Department of Gerontology, and Geriatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China;
| | - Yang Li
- Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang 110004, China;
| | - Te Qi
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang 110004, China; (Z.Z.); (T.Q.); (Y.Q.); (Y.H.); (Z.L.)
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Departments of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ying Qi
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang 110004, China; (Z.Z.); (T.Q.); (Y.Q.); (Y.H.); (Z.L.)
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Departments of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yujing Huang
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang 110004, China; (Z.Z.); (T.Q.); (Y.Q.); (Y.H.); (Z.L.)
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Departments of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Zhongyang Liu
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang 110004, China; (Z.Z.); (T.Q.); (Y.Q.); (Y.H.); (Z.L.)
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Departments of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Qiang Ruan
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang 110004, China; (Z.Z.); (T.Q.); (Y.Q.); (Y.H.); (Z.L.)
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Departments of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yanping Ma
- Virology Laboratory, Shengjing Hospital of China Medical University, Shenyang 110004, China; (Z.Z.); (T.Q.); (Y.Q.); (Y.H.); (Z.L.)
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Departments of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
33
|
Cervera-Juanes R, Zimmerman KD, Wilhelm L, Zhu D, Bodie J, Kohama SG, Urbanski HF. Modulation of neural gene networks by estradiol in old rhesus macaque females. GeroScience 2024; 46:5819-5841. [PMID: 38509416 PMCID: PMC11493911 DOI: 10.1007/s11357-024-01133-z] [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: 01/08/2024] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
The postmenopausal decrease in circulating estradiol (E2) levels has been shown to contribute to several adverse physiological and psychiatric effects. To elucidate the molecular effects of E2 on the brain, we examined differential gene expression and DNA methylation (DNAm) patterns in the nonhuman primate brain following ovariectomy (Ov) and subsequent subcutaneous bioidentical E2 chronic treatment. We identified several dysregulated molecular networks, including MAPK signaling and dopaminergic synapse response, that are associated with ovariectomy and shared across two different brain areas, the occipital cortex (OC) and prefrontal cortex (PFC). The finding that hypomethylation (p = 1.6 × 10-51) and upregulation (p = 3.8 × 10-3) of UBE2M across both brain regions provide strong evidence for molecular differences in the brain induced by E2 depletion. Additionally, differential expression (p = 1.9 × 10-4; interaction p = 3.5 × 10-2) of LTBR in the PFC provides further support for the role E2 plays in the brain, by demonstrating that the regulation of some genes that are altered by ovariectomy may also be modulated by Ov followed by hormone replacement therapy (HRT). These results present real opportunities to understand the specific biological mechanisms that are altered with depleted E2. Given E2's potential role in cognitive decline and neuroinflammation, our findings could lead to the discovery of novel therapeutics to slow cognitive decline. Together, this work represents a major step toward understanding molecular changes in the brain that are caused by ovariectomy and how E2 treatment may revert or protect against the negative neuro-related consequences caused by a depletion in estrogen as women approach menopause.
Collapse
Affiliation(s)
- Rita Cervera-Juanes
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
- Center for Precision Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
| | - Kip D Zimmerman
- Center for Precision Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Larry Wilhelm
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Dongqin Zhu
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Jessica Bodie
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Steven G Kohama
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
| | - Henryk F Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| |
Collapse
|
34
|
Nasser M, Abd El-Latif MB, Mahmoud A, Diaa D, Kamal G, Mahmoud H, Emad M, Hany M, Hany R, Mohamed S, Farid A. Utilization of Ziziphus spina-christi leaf extract-loaded chitosan nanoparticles in wastewater treatment and their impact on animal health. Int J Biol Macromol 2024; 282:137441. [PMID: 39522915 DOI: 10.1016/j.ijbiomac.2024.137441] [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] [Revised: 10/13/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Ziziphus leaf extract (ZEX), chitosan nanoparticles (CS NPs) and Ziziphus leaf extract loaded chitosan nanoparticles (ZEX-CS NPs) were prepared in this study and after chemical analysis and characterization, they were used in wastewater purification. The study also aimed to establish, using an animal model, the feasibility of employing treated water in drinking applications. ZEX-CS NPs were prepared by ionic gelation method. About 25 male Sprague Dawley rats (10 weeks and 170-200 g) were divided into five groups (5 rats/group): group I received tape water; group II received untreated wastewater, group III received ZEX treated wastewater, group IV received CS NPs treated wastewater and group V received ZEX-CS NPs treated wastewater. ZEX-CS NPs have a size of 73 nm, hydrodynamic size of 85.81 nm and zeta potential of -33.68 mV. In addition, ZEX-CS NPs have stronger antioxidant and anti-inflammatory activity with moderate anti-coagulant activity and weaker cytotoxicity than ZEX and CS NPs. Group II showed a significant elevation in the kidney function parameters, oxidative stress and cytokine levels when compared to the other groups, in addition; no significant differences were found in all measured parameters between the rats of group I and V. ZEX-CS NPs were effective in wastewater purification.
Collapse
Affiliation(s)
- Mohamed Nasser
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt.
| | - Mona B Abd El-Latif
- Environmental Research Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Alaa Mahmoud
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Dai Diaa
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Germeen Kamal
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Hagar Mahmoud
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Merna Emad
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Merna Hany
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Rawan Hany
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Sohaila Mohamed
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Alyaa Farid
- Biotechnology Department, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
35
|
Mocarski ES. Cytomegalovirus Biology Viewed Through a Cell Death Suppression Lens. Viruses 2024; 16:1820. [PMID: 39772130 PMCID: PMC11680106 DOI: 10.3390/v16121820] [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: 10/28/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Cytomegaloviruses, species-specific members of the betaherpesviruses, encode an impressive array of immune evasion strategies committed to the manipulation of the host immune system enabling these viruses to remain for life in a stand-off with host innate and adaptive immune mechanisms. Even though they are species-restricted, cytomegaloviruses are distributed across a wide range of different mammalian species in which they cause systemic infection involving many different cell types. Regulated, or programmed cell death has a recognized potential to eliminate infected cells prior to completion of viral replication and release of progeny. Cell death also naturally terminates replication during the final stages of replication. Over the past two decades, the host defense potential of known programmed cell death pathways (apoptosis, necroptosis, and pyroptosis), as well as a novel mitochondrial serine protease pathway have been defined through studies of cytomegalovirus-encoded cell death suppressors. Such virus-encoded inhibitors prevent virus-induced, cytokine-induced, and stress-induced death of infected cells while also moderating inflammation. By evading cell death and consequent inflammation as well as innate and adaptive immune clearance, cytomegaloviruses represent successful pathogens that become a critical disease threat when the host immune system is compromised. This review will discuss cell death programs acquired for mammalian host defense against cytomegaloviruses and enumerate the range of modulatory strategies this type of virus employs to balance host defense in favor of lifelong persistence.
Collapse
Affiliation(s)
- Edward S. Mocarski
- Department of Microbiology & Immunology, Stanford Medical School, Stanford University, Stanford, CA 94305, USA;
- Department of Microbiology & Immunology, Emory Medical School, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
36
|
Zhang Y, Wu R, Zhan X, Wang XY, Xiang LW, Duan YQ, You Y, Zhang JB, Wu R, Zhang YY, Duan L. Neutrophil extracellular traps facilitate liver inflammation/fibrosis progression by entering macrophages and triggering AIM2 inflammasome-dependent pyroptosis. Cell Commun Signal 2024; 22:556. [PMID: 39568027 PMCID: PMC11577833 DOI: 10.1186/s12964-024-01944-9] [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: 08/21/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Absent in melanoma 2 (AIM2) inflammasome-dependent pyroptosis and neutrophil extracellular traps (NETs) have been implicated in chronic liver disease (CLD). However, the specific intrahepatic cell type that undergoes AIM2 inflammasome-dependent pyroptosis and how their interaction augments hepatic inflammation/fibrosis remains unclear. METHODS The expression and correlation of AIM2 inflammasome-dependent pyroptosis-related indicators and NETs were analyzed in biopsy tissue and blood specimens from chronic hepatitis patients (CHs). Cell-based experiments were conducted to investigate their interaction. In vitro and in vivo experiments were used to analyze their effects on the progression of hepatic inflammation/fibrosis as well as their clinical importance. RESULTS Elevated levels of AIM2 inflammasome-dependent pyroptosis indicators and NETs were detected in biopsy tissue and blood specimens. Circulating NETs were positively correlated with pyroptosis-related indicators, and both were related with disease severity. Confocal imaging revealed that AIM2 was mainly localized to hepatic macrophages, indicating that hepatic macrophages were the major cell type that underwent pyroptosis. NETs were directly engulfed by macrophages and then stimulated AIM2 inflammasome-dependent macrophage pyroptosis in vitro, which amplified the activation of hepatic stellate cells (HSCs) and increased collagen deposition. Administration of the NETs degradation agent DNase I or the AIM2 inflammasome activation inhibitor ODN A151 effectively alleviated chronic liver inflammation/fibrosis progression in vivo. CONCLUSIONS NETs-induced AIM2 inflammasome-dependent pyroptosis in macrophages facilitates liver inflammation/fibrosis progression. The identified NET-AIM2 inflammasome cascade could serve as a novel therapeutic target for hepatic inflammation/fibrosis progression.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, No. 74 Lin Jiang Road, Chongqing, 400010, China
| | - Rong Wu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, No. 74 Lin Jiang Road, Chongqing, 400010, China
| | - Xi Zhan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, No. 74 Lin Jiang Road, Chongqing, 400010, China
| | - Xuan-Yi Wang
- Department of Pathology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin-Wei Xiang
- Department of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Ya-Qian Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, No. 74 Lin Jiang Road, Chongqing, 400010, China
| | - Yan You
- Department of Pathology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian-Bo Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Rui Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1 You Yi Road, Chongqing, 400016, China.
| | - Yun-Yuan Zhang
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, No.16 Jiang Su Road, Qingdao, 266003, China.
| | - Liang Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, No. 74 Lin Jiang Road, Chongqing, 400010, China.
| |
Collapse
|
37
|
Lin C, Zhang C, Chen N, Meurens F, Zhu J, Zheng W. How Does African Swine Fever Virus Evade the cGAS-STING Pathway? Pathogens 2024; 13:957. [PMID: 39599510 PMCID: PMC11597325 DOI: 10.3390/pathogens13110957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
African swine fever (ASF), a highly infectious and devastating disease affecting both domestic pigs and wild boars, is caused by the African swine fever virus (ASFV). ASF has resulted in rapid global spread of the disease, leading to significant economic losses within the swine industry. A significant obstacle to the creation of safe and effective ASF vaccines is the existing knowledge gap regarding the pathogenesis of ASFV and its mechanisms of immune evasion. The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway is a major pathway mediating type I interferon (IFN) antiviral immune response against infections by diverse classes of pathogens that contain DNA or generate DNA in their life cycles. To evade the host's innate immune response, ASFV encodes many proteins that inhibit the production of type I IFN by antagonizing the cGAS-STING signaling pathway. Multiple proteins of ASFV are involved in promoting viral replication by protein-protein interaction during ASFV infection. The protein QP383R could impair the function of cGAS. The proteins EP364R, C129R and B175L could disturb the function of cyclic guanosine monophosphate-adenosine monophosphate (cGAMP). The proteins E248R, L83L, MGF505-11L, MGF505-7R, H240R, CD2v, E184L, B175L and p17 could interfere with the function of STING. The proteins MGF360-11L, MGF505-7R, I215L, DP96R, A151R and S273R could affect the function of TANK Binding Kinase 1 (TBK1) and IκB kinase ε (IKKε). The proteins MGF360-14L, M1249L, E120R, S273R, D129L, E301R, DP96R, MGF505-7R and I226R could inhibit the function of Interferon Regulatory Factor 3 (IRF3). The proteins MGF360-12L, MGF505-7R/A528R, UBCv1 and A238L could inhibit the function of nuclear factor kappa B (NF-Κb).
Collapse
Affiliation(s)
- Can Lin
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (C.L.); (C.Z.); (N.C.); (J.Z.)
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Chenyang Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (C.L.); (C.Z.); (N.C.); (J.Z.)
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Nanhua Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (C.L.); (C.Z.); (N.C.); (J.Z.)
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - François Meurens
- Swine and Poultry Infectious Diseases Research Center, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada;
- Department of Veterinary Microbiology and Immunology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Jianzhong Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (C.L.); (C.Z.); (N.C.); (J.Z.)
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Wanglong Zheng
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (C.L.); (C.Z.); (N.C.); (J.Z.)
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| |
Collapse
|
38
|
Akcay E, Karatas H. P2X7 receptors from the perspective of NLRP3 inflammasome pathway in depression: Potential role of cannabidiol. Brain Behav Immun Health 2024; 41:100853. [PMID: 39296605 PMCID: PMC11407962 DOI: 10.1016/j.bbih.2024.100853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/16/2024] [Accepted: 09/02/2024] [Indexed: 09/21/2024] Open
Abstract
Many patients with depressive disorder do not respond to conventional antidepressant treatment. There is an ongoing interest in investigating potential mechanisms of treatment resistance in depression to provide alternative treatment options involving inflammatory mechanisms. Increasing evidence implicates the NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome as a critical factor in neuroinflammation. ATP-induced P2X7 receptor (P2X7R) activation is a major trigger for inflammation, activating the canonical NLRP3 inflammatory cascade. Psychosocial stress, the primary environmental risk factor for depression, is associated with changes in ATP-mediated P2X7R signaling. Depression and stress response can be alleviated by Cannabidiol (CBD). CBD has an anti-inflammatory activity related to the regulation of NLRP3 inflammasome activation. However, CBD's effects on the inflammasome pathway are poorly understood in central nervous system (CNS) cells, including microglia, astrocytes, and neurons. This review will emphasize some findings for neuroinflammation and NLRP3 inflammasome pathway involvement in depression, particularly addressing the ATP-induced P2X7R activation. Moreover, we will underline evidence for the effect of CBD on depression and address its potential impacts on neuroinflammation through the NLRP3 inflammasome cascade.
Collapse
Affiliation(s)
- Elif Akcay
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey
- University of Health Sciences, Ankara Bilkent City Hospital, Department of Child and Adolescent Psychiatry, Ankara, Turkey
| | - Hulya Karatas
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey
| |
Collapse
|
39
|
Paduraru E, Jijie R, Simionov IA, Gavrilescu CM, Ilie T, Iacob D, Lupitu A, Moisa C, Muresan C, Copolovici L, Copolovici DM, Mihalache G, Lipsa FD, Solcan G, Danelet GA, Nicoara M, Ciobica A, Solcan C. Honey Enriched with Additives Alleviates Behavioral, Oxidative Stress, and Brain Alterations Induced by Heavy Metals and Imidacloprid in Zebrafish. Int J Mol Sci 2024; 25:11730. [PMID: 39519279 PMCID: PMC11546825 DOI: 10.3390/ijms252111730] [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: 09/27/2024] [Revised: 10/22/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Environmental concerns have consistently been a focal point for the scientific community. Pollution is a critical ecological issue that poses significant threats to human health and agricultural production. Contamination with heavy metals and pesticides is a considerable concern, a threat to the environment, and warrants special attention. In this study, we investigated the significant issues arising from sub-chronic exposure to imidacloprid (IMI), mercury (Hg), and cadmium (Cd), either alone or in combination, using zebrafish (Danio rerio) as an animal model. Additionally, we assessed the potential protective effects of polyfloral honey enriched with natural ingredients, also called honey formulation (HF), against the combined sub-chronic toxic effects of the three contaminants. The effects of IMI (0.5 mg·L-1), Hg (15 μg·L-1), and Cd (5 μg·L-1), both individually and in combination with HF (500 mg·L-1), on zebrafish were evaluated by quantifying acetylcholinesterase (AChE) activity, lipid peroxidation (MDA), various antioxidant enzyme activities like superoxide dismutase and glutathione peroxidase (SOD and GPx), 2D locomotor activity, social behavior, histological and immunohistochemical factors, and changes in body element concentrations. Our findings revealed that all concentrations of pollutants may disrupt social behavior, diminish swimming performances (measured by total distance traveled, inactivity, and swimming speed), and elevate oxidative stress (OS) biomarkers of SOD, GPx, and MDA in zebrafish over the 21-day administration period. Fish exposed to IMI and Hg + Cd + IMI displayed severe lesions and increased GFAP (Glial fibrillary acidic protein) and S100B (S100 calcium-binding protein B) protein expression in the optic tectum and cerebellum, conclusively indicating astrocyte activation and neurotoxic effects. Furthermore, PCNA (Proliferating cell nuclear antigen) staining revealed reduced cell proliferation in the IMI-exposed group, contrasting with intensified proliferation in the Hg + Cd group. The nervous system exhibited significant damage across all studied concentrations, confirming the observed behavioral changes. Moreover, HF supplementation significantly mitigated the toxicity induced by contaminants and reduced OS. Therefore, the exposure to chemical mixtures offers a more complete picture of adverse impacts on aquatic ecosystems and the supplementation with bioactive compounds can help to reduce the toxicity induced by exposure to environmental pollutants.
Collapse
Affiliation(s)
- Emanuela Paduraru
- Doctoral School of Geosciences, Faculty of Geography and Geology, Alexandru Ioan Cuza University of Iasi, No. 20 A Carol I Avenue, 700505 Iasi, Romania; (E.P.); (D.I.); (M.N.)
| | - Roxana Jijie
- Research Center on Advanced Materials and Technologies (RAMTECH), Department of Exact and Natural Sciences, Institute of Interdisciplinary Research, Alexandru Ioan Cuza University of Iasi, No. 11 Carol I Avenue, 700506 Iasi, Romania;
| | - Ira-Adeline Simionov
- Department of Food Science, Food Engineering, Biotechnologies and Aquaculture, Dunarea de Jos University of Galati, No. 47 Domnească Street, 800008 Galati, Romania;
- REXDAN Research Infrastructure, Dunarea de Jos University of Galati, No. 98 George Coșbuc Street, 800385 Galati, Romania
| | - Cristina-Maria Gavrilescu
- Department of Biomedical Sciences, Grigore T. Popa University of Medicine and Pharmacy, No. 16 University Street, 700115 Iasi, Romania;
| | - Tudor Ilie
- Synergy Plant Products, No. 12 Milano Street, Prejmer, 507165 Brasov, Romania;
| | - Diana Iacob
- Doctoral School of Geosciences, Faculty of Geography and Geology, Alexandru Ioan Cuza University of Iasi, No. 20 A Carol I Avenue, 700505 Iasi, Romania; (E.P.); (D.I.); (M.N.)
| | - Andreea Lupitu
- Faculty of Food Engineering, Tourism and Environmental Protection, Institute for Research, Development and Innovation in Technical and Natural Sciences, Aurel Vlaicu University, No. 2 Elena Dragoi Street, 310330 Arad, Romania; (A.L.); (C.M.); (C.M.); (L.C.); (D.M.C.)
| | - Cristian Moisa
- Faculty of Food Engineering, Tourism and Environmental Protection, Institute for Research, Development and Innovation in Technical and Natural Sciences, Aurel Vlaicu University, No. 2 Elena Dragoi Street, 310330 Arad, Romania; (A.L.); (C.M.); (C.M.); (L.C.); (D.M.C.)
| | - Claudia Muresan
- Faculty of Food Engineering, Tourism and Environmental Protection, Institute for Research, Development and Innovation in Technical and Natural Sciences, Aurel Vlaicu University, No. 2 Elena Dragoi Street, 310330 Arad, Romania; (A.L.); (C.M.); (C.M.); (L.C.); (D.M.C.)
| | - Lucian Copolovici
- Faculty of Food Engineering, Tourism and Environmental Protection, Institute for Research, Development and Innovation in Technical and Natural Sciences, Aurel Vlaicu University, No. 2 Elena Dragoi Street, 310330 Arad, Romania; (A.L.); (C.M.); (C.M.); (L.C.); (D.M.C.)
| | - Dana M. Copolovici
- Faculty of Food Engineering, Tourism and Environmental Protection, Institute for Research, Development and Innovation in Technical and Natural Sciences, Aurel Vlaicu University, No. 2 Elena Dragoi Street, 310330 Arad, Romania; (A.L.); (C.M.); (C.M.); (L.C.); (D.M.C.)
| | - Gabriela Mihalache
- Integrated Center of Environmental Science Studies in the North-Eastern Development Region (CERNESIM), Department of Exact and Natural Sciences, Institute of Interdisciplinary Research, Alexandru Ioan Cuza University of Iasi, No. 11 Carol I Avenue, 700506 Iasi, Romania;
| | - Florin Daniel Lipsa
- Department of Food Technologies, Ion Ionescu de la Brad University of Life Sciences, No. 3 Mihail Sadoveanu Alley, 700490 Iasi, Romania;
| | - Gheorghe Solcan
- Faculty of Veterinary Medicine, Ion Ionescu de la Brad University of Life Sciences, No. 8 Mihail Sadoveanu Alley, 700489 Iasi, Romania; (G.S.); (G.-A.D.); (C.S.)
| | - Gabriela-Alexandra Danelet
- Faculty of Veterinary Medicine, Ion Ionescu de la Brad University of Life Sciences, No. 8 Mihail Sadoveanu Alley, 700489 Iasi, Romania; (G.S.); (G.-A.D.); (C.S.)
| | - Mircea Nicoara
- Doctoral School of Geosciences, Faculty of Geography and Geology, Alexandru Ioan Cuza University of Iasi, No. 20 A Carol I Avenue, 700505 Iasi, Romania; (E.P.); (D.I.); (M.N.)
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, No. 20A Carol I Avenue, 700505 Iasi, Romania
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, No. 20A Carol I Avenue, 700505 Iasi, Romania
- Center of Biomedical Research, Romanian Academy, No. 8 Carol I Avenue, 700506 Iasi, Romania
- Academy of Romanian Scientists, No. 54 Independence Street, Sector 5, 050094 Bucharest, Romania
- “Ioan Haulica” Institute, Apollonia University, No. 11 Pacurari Street, 700511 Iasi, Romania
| | - Carmen Solcan
- Faculty of Veterinary Medicine, Ion Ionescu de la Brad University of Life Sciences, No. 8 Mihail Sadoveanu Alley, 700489 Iasi, Romania; (G.S.); (G.-A.D.); (C.S.)
| |
Collapse
|
40
|
Bouwman W, Verhaegh W, van Doorn A, Raymakers R, van der Poll T, van de Stolpe A. Quantitative characterization of immune cells by measuring cellular signal transduction pathway activity. Sci Rep 2024; 14:24487. [PMID: 39424625 PMCID: PMC11489675 DOI: 10.1038/s41598-024-75666-w] [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: 03/27/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024] Open
Abstract
For many diseases, including cancer, infections, and auto-immune diseases, the immune response is a major determinant of disease progression, response to therapy, and clinical outcome. Innate and adaptive immune responses are controlled by coordinated activity of different immune cell types. The functional activity state of immune cells is determined by Signal Transduction Pathways (STPs). A recently developed technology (Simultaneous Transcriptome-based Activity Profiling of Signal Transduction Pathways, STAP-STP) enables simultaneous and quantitative activity measurement of relevant STPs in immune cells based on mRNA-analysis. STAP-STP technology was used to analyze public transcriptome data of a variety of immune cell types in resting and activated functional state. In addition, a clinical study on rheumatoid arthritis (RA) was analyzed to illustrate utility of the technology. Per sample, activity of androgen and estrogen receptor, PI3K, MAPK, TGFβ, Notch, NFκB, JAK-STAT1/2, and JAK-STAT3 STPs was calculated, generating an STP activity profile (SAP) consisting of 9 activity scores. Each analyzed immune cell type, i.e. naive/resting and immune-activated CD4 + and CD8 + T cells, T helper cells, B cells, NK cells, monocytes, macrophages, and dendritic cells, had a reproducible and characteristic SAP, reflecting both cell type and its activity state. Analysis of clinical RA samples revealed increased TGFβ STP activity in whole blood samples. In conclusion, STAP-STP technology enables quantitative measurement of the functional activity state of immune cells of the innate and adaptive immune system. Aside from diagnostic applications, utility lies in unravelling abnormal immune function in disease and immunomodulatory drug development.
Collapse
Affiliation(s)
- Wilbert Bouwman
- Center of Experimental and Molecular Medicine & Division of Infectious Diseases, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | - Tom van der Poll
- Center of Experimental and Molecular Medicine & Division of Infectious Diseases, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
41
|
Wang J, Yang J, Narang A, He J, Wolfgang C, Li K, Zheng L. Consensus, debate, and prospective on pancreatic cancer treatments. J Hematol Oncol 2024; 17:92. [PMID: 39390609 PMCID: PMC11468220 DOI: 10.1186/s13045-024-01613-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Pancreatic cancer remains one of the most aggressive solid tumors. As a systemic disease, despite the improvement of multi-modality treatment strategies, the prognosis of pancreatic cancer was not improved dramatically. For resectable or borderline resectable patients, the surgical strategy centered on improving R0 resection rate is consensus; however, the role of neoadjuvant therapy in resectable patients and the optimal neoadjuvant therapy of chemotherapy with or without radiotherapy in borderline resectable patients were debated. Postoperative adjuvant chemotherapy of gemcitabine/capecitabine or mFOLFIRINOX is recommended regardless of the margin status. Chemotherapy as the first-line treatment strategy for advanced or metastatic patients included FOLFIRINOX, gemcitabine/nab-paclitaxel, or NALIRIFOX regimens whereas 5-FU plus liposomal irinotecan was the only standard of care second-line therapy. Immunotherapy is an innovative therapy although anti-PD-1 antibody is currently the only agent approved by for MSI-H, dMMR, or TMB-high solid tumors, which represent a very small subset of pancreatic cancers. Combination strategies to increase the immunogenicity and to overcome the immunosuppressive tumor microenvironment may sensitize pancreatic cancer to immunotherapy. Targeted therapies represented by PARP and KRAS inhibitors are also under investigation, showing benefits in improving progression-free survival and objective response rate. This review discusses the current treatment modalities and highlights innovative therapies for pancreatic cancer.
Collapse
Affiliation(s)
- Junke Wang
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jie Yang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Amol Narang
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jin He
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Christopher Wolfgang
- Department of Surgery, New York University School of Medicine and NYU-Langone Medical Center, New York, NY, USA
| | - Keyu Li
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA.
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Lei Zheng
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA.
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- The Multidisciplinary Gastrointestinal Cancer Laboratories Program, the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
42
|
Zhang Z, Su M, Jiang P, Wang X, Tong X, Wu G. Unlocking Apoptotic Pathways: Overcoming Tumor Resistance in CAR-T-Cell Therapy. Cancer Med 2024; 13:e70283. [PMID: 39377542 PMCID: PMC11459502 DOI: 10.1002/cam4.70283] [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: 07/01/2024] [Revised: 09/03/2024] [Accepted: 09/20/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-T-cell therapy has transformed cancer treatment, leading to remarkable clinical outcomes. However, resistance continues to be a major obstacle, significantly limiting its efficacy in numerous patients. OBJECTIVES This review critically examines the challenges associated with CAR-T-cell therapy, with a particular focus on the role of apoptotic pathways in overcoming resistance. METHODS We explore various strategies to sensitize tumor cells to CAR-T-cell-mediated apoptosis, including the use of combination therapies with BH3 mimetics, Mcl-1 inhibitors, IAP inhibitors, and HDAC inhibitors. These agents inhibit anti-apoptotic proteins and activate intrinsic mitochondrial pathways, enhancing the susceptibility of tumor cells to apoptosis. Moreover, targeting the extrinsic pathway can increase the expression of death receptors on tumor cells, further promoting their apoptosis. The review also discusses the development of novel CAR constructs that enhance anti-apoptotic protein expression, such as Bcl-2, which may counteract CAR-T cell exhaustion and improve antitumor efficacy. We assess the impact of the tumor microenvironment (TME) on CAR-T cell function and propose dual-targeting CAR-T cells to simultaneously address both myeloid-derived suppressor cells (MDSCs) and tumor cells. Furthermore, we explore the potential of combining agents like PPAR inhibitors to activate the cGAS-STING pathway, thereby improving CAR-T cell infiltration into the tumor. CONCLUSIONS This review highlights that enhancing tumor cell sensitivity to apoptosis and increasing CAR-T cell cytotoxicity through apoptotic pathways could significantly improve therapeutic outcomes. Targeting apoptotic proteins, particularly those involved in the intrinsic mitochondrial pathway, constitutes a novel approach to overcoming resistance. The insights presented herein lay a robust foundation for future research and clinical applications aimed at optimizing CAR-T cell therapies.
Collapse
Affiliation(s)
- Zhanna Zhang
- Department of HematologyDongyang Hospital Affiliated to WenZhou Medical UniversityJinhuaZhejiangChina
| | - Manqi Su
- Department of HematologyDongyang Hospital Affiliated to WenZhou Medical UniversityJinhuaZhejiangChina
| | - Panruo Jiang
- Department of HematologyDongyang Hospital Affiliated to WenZhou Medical UniversityJinhuaZhejiangChina
| | - Xiaoxia Wang
- Department of HematologyDongyang Hospital Affiliated to WenZhou Medical UniversityJinhuaZhejiangChina
| | - Xiangmin Tong
- Department of Central LaboratorySchool of Medicine, Affiliated Hangzhou First People's Hospital, WestLake UniversityZhejiangHangzhouChina
| | - Gongqiang Wu
- Department of HematologyDongyang Hospital Affiliated to WenZhou Medical UniversityJinhuaZhejiangChina
| |
Collapse
|
43
|
Grabiec M, Sobstyl M, Skirecki T. Nod-like receptors: The relevant elements of glioblastoma`s prognostic puzzle. Pharmacol Res 2024; 208:107411. [PMID: 39270948 DOI: 10.1016/j.phrs.2024.107411] [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: 03/28/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
Despite considerable improvements in understanding the biology of glioblastoma (GB), it still remains the most lethal type of brain tumor in adults. The role of innate immune cells in the development of GB was recently described. In particular, the tumor-immune cell interactions are thought to be critical in enabling tumor tolerance and even protection against therapeutics. Interestingly, the GB cells express proteins belonging to the family of intracellular pattern-recognition receptors, namely the NOD-like receptors (NLRs). Their activation may trigger the formation of the inflammasome complex leading to the secretion of mature IL-1β and IL-18 and thus resulting in cell death. Intrudingly, the expression of most NLRs was found to be correlated with tumor progression and poor prognosis. We speculate that recognizing the role of NOD-like receptors in GB has the potential to improve the effectiveness of diagnostic tools and prognosis, while also encouraging the development of novel precision medicine-based therapies.
Collapse
Affiliation(s)
- Marta Grabiec
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland.
| | - Michał Sobstyl
- Department of Neurosurgery, Institute of Psychiatry and Neurology, Warsaw, Poland
| | - Tomasz Skirecki
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| |
Collapse
|
44
|
Yan W, Wang C, Zhao Y, Jiang Y, Sun M. Involvement of Calpain in Neurovascular Unit Damage through Up-regulating PARP-NF-κB Signaling during Experimental Ischemic Stroke. Mol Neurobiol 2024; 61:8104-8122. [PMID: 38472651 DOI: 10.1007/s12035-024-04092-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 03/03/2024] [Indexed: 03/14/2024]
Abstract
Calpain and PARP-NF-κB signaling are reported to participate in the ischemic brain injury. In this study, it was investigated whether calpain was contributed to the neurovascular unit (NVU) damage through up-regulating PARP-NF-κB signaling during experimental ischemic stroke. Male Sprague-Dawley rats were suffered from 90 min of middle cerebral artery occlusion, followed by reperfusion. The NVU damage was evaluated by the permeability of blood-brain barrier (BBB), the degradation of proteins in extracellular matrix and tight junctions, and ultrastructural changes. The inflammatory response was determined by the expression of inflammatory genes driven by PARP-NF-κB signaling and the activities of myeloperoxidase (MPO). Treatment with MDL 28,170, a calpain inhibitor, improved neurological functions, reduced TUNEL staining index, lessened brain swelling, and decreased infarct volume in ischemic rats. Moreover, it reduced the BBB permeability, enhanced the levels of laminin, collagen IV and occludin, and attenuated the ultrastructural damage of NVU in penumbra and core after induction of ischemia. Meanwhile, it enhanced the levels of cytosolic IκBα, lessened the levels of nuclear PARP and NF-κB p65, reduced the levels of ICAM-1, TNF-α, IL-1β, MMP-9, and MMP-2,and suppressed the activities of MPO in penumbra and core. These data showed that calpain inhibition suppressed PARP-NF-κB signaling-mediated inflammatory response, reduced NVU damage, and protected brain against ischemic stroke, suggesting the involvement of calpain in the NVU damage through up-regulating PARP-NF-κB signaling during brain ischemia.
Collapse
Affiliation(s)
- Wenhao Yan
- Department of Pediatrics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Chunyang Wang
- Department of Neuropharmacology, Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Yumei Zhao
- Department of Neuropharmacology, Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Yingying Jiang
- Department of Neuropharmacology, Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Ming Sun
- Department of Neuropharmacology, Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China.
| |
Collapse
|
45
|
Aalto AL, Luukkonen V, Meinander A. Ubiquitin signalling in Drosophila innate immune responses. FEBS J 2024; 291:4397-4413. [PMID: 38069549 DOI: 10.1111/febs.17028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/24/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
Cells respond to invading pathogens and danger signals from the environment by adapting gene expression to meet the need for protective effector molecules. While this innate immune response is required for the cell and the organism to recover, excess immune activation may lead to loss of homeostasis, thereby promoting chronic inflammation and cancer progression. The molecular basis of innate immune defence is comprised of factors promoting survival and proliferation, such as cytokines, antimicrobial peptides and anti-apoptotic proteins. As the molecular mechanisms regulating innate immune responses are conserved through evolution, the fruit fly Drosophila melanogaster serves as a convenient, affordable and ethical model organism to enhance understanding of immune signalling. Fly immunity against bacterial infection is built up by both cellular and humoral responses, where the latter is regulated by the Imd and Toll pathways activating NF-κB transcription factors Relish, Dorsal and Dif, as well as JNK activation and JAK/STAT signalling. As in mammals, the Drosophila innate immune signalling pathways are characterised by ubiquitination of signalling molecules followed by ubiquitin receptors binding to the ubiquitin chains, as well as by rapid changes in protein levels by ubiquitin-mediated targeted proteasomal and lysosomal degradation. In this review, we summarise the molecular signalling pathways regulating immune responses to pathogen infection in Drosophila, with a focus on ubiquitin-dependent control of innate immunity and inflammatory signalling.
Collapse
Affiliation(s)
- Anna L Aalto
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, Åbo Akademi University, Turku, Finland
| | - Veera Luukkonen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | - Annika Meinander
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, Åbo Akademi University, Turku, Finland
| |
Collapse
|
46
|
Zibandeh N, Li Z, Ogg G, Bottomley MJ. Cutaneous adaptive immunity and uraemia: a narrative review. Front Immunol 2024; 15:1464338. [PMID: 39399503 PMCID: PMC11466824 DOI: 10.3389/fimmu.2024.1464338] [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: 07/13/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024] Open
Abstract
Chronic kidney disease affects 1 in 10 people globally, with a prevalence twenty times that of cancer. A subset of individuals will progress to end-stage renal disease (ESRD) where renal replacement therapy is required to maintain health. Cutaneous disease, including xerosis and pruritus, are endemic amongst patients with ESRD. In the uraemia-associated immune deficiency of ESRD, impaired circulating immune responses contribute to increased infection risk and poorer vaccination response. Clinical manifestations of dysregulated adaptive immunity within the skin have been well-described and have been posited to play a role in cutaneous features of ESRD. However, our understanding of the mechanisms by which adaptive immunity within the skin is affected by uraemia is relatively limited. We provide an overview of how the cutaneous adaptive immune system is impacted both directly and indirectly by uraemia, highlighting that much work has been extrapolated from the circulating immune system and often has not been directly evaluated in the skin compartment. We identify knowledge gaps which may be addressed by future research. Ultimately, greater understanding of these pathways may facilitate novel therapeutic approaches to ameliorate widespread cutaneous symptomatology in ESRD.
Collapse
Affiliation(s)
- Noushin Zibandeh
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, United Kingdom
| | - Zehua Li
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, United Kingdom
| | - Graham Ogg
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, United Kingdom
- Department of Dermatology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- MRC Translational Immune Discovery Unit , University of Oxford, Oxford, United Kingdom
| | - Matthew J. Bottomley
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, United Kingdom
- Oxford Kidney and Transplant Unit, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
47
|
Stocks H, De Leeuw E, Lambrecht BN, Vandekerckhove L, van Loo G, Wullaert A. Development of human innate immune responses in a humanized mouse model expressing four human myelopoiesis transgenes. Front Immunol 2024; 15:1419117. [PMID: 39399507 PMCID: PMC11466769 DOI: 10.3389/fimmu.2024.1419117] [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: 04/17/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
Background Dysregulated innate immune responses underlie multiple inflammatory diseases, but clinical translation of preclinical innate immunity research in mice is hampered by the difficulty of studying human inflammatory reactions in an in vivo context. We therefore sought to establish in vivo human inflammatory responses in NSG-QUAD mice that express four human myelopoiesis transgenes to improve engraftment of a human innate immune system. Methods We reconstituted NSG-QUAD mice with human hematopoietic stem and progenitor cells (HSPCs), after which we evaluated human myeloid cell development and subsequent human responses to systemic and local lipopolysaccharide (LPS) challenges. Results NSG-QUAD mice already displayed engraftment of human monocytes, dendritic cells and granulocytes in peripheral blood, spleen and liver at 6 weeks after HSPC reconstitution, in which both classical, intermediate and non-classical monocytes were present. These huNSG-QUAD mice responded to intraperitoneal and intranasal LPS challenges with production of NF-κB-dependent human cytokines, a human type I interferon response, as well as inflammasome-mediated production of human IL-1β and IL-18. The latter were specifically abrogated by the NLRP3 inhibitor MCC950, while LPS-induced human monocyte death was not altered. Besides providing proof-of-principle for small molecule testing of human inflammatory reactions in huNSG-QUAD mice, this observation suggests that LPS-induced in vivo release of human NLRP3 inflammasome-generated cytokines occurs in a cell death-independent manner. Conclusion HuNSG-QUAD mice are competent for the NF-κB, interferon and inflammasome effectors of human innate immunity, and can thus be utilized to investigate signaling mechanisms and pharmacological targeting of human inflammatory responses in an in vivo setting.
Collapse
Affiliation(s)
- Hannah Stocks
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
| | - Elisabeth De Leeuw
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
| | - Bart N Lambrecht
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
| | - Linos Vandekerckhove
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- HIV Cure and Research Center (HCRC), Ghent, Belgium
| | - Geert van Loo
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Andy Wullaert
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Cell Death Signaling Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
48
|
Nasr S, Dawood AS, Ibrahim AM, Abdel-Aziz MS, Fayad W, Abdelnaser A, El-Hady FKA. Anti-inflammatory potential of aspergillus unguis SP51-EGY: TLR4-dependent effects & chemical diversity via Q-TOF LC-HRMS. BMC Biotechnol 2024; 24:62. [PMID: 39294631 PMCID: PMC11411751 DOI: 10.1186/s12896-024-00890-1] [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: 10/06/2023] [Accepted: 08/28/2024] [Indexed: 09/21/2024] Open
Abstract
Inflammation serves as an intricate defense mechanism for tissue repair. However, overactivation of TLR4-mediated inflammation by lipopolysaccharide (LPS) can lead to detrimental outcomes such as sepsis, acute lung injury, and chronic inflammation, often associated with cancer and autoimmune diseases. This study delves into the anti-inflammatory properties of "Aspergillus unguis isolate SP51-EGY" on LPS-stimulated RAW 264.7 macrophages. Through real-time qPCR, we assessed the expression levels of pivotal inflammatory genes, including iNOS, COX-2, TNF-α, and IL-6. Remarkably, our fungal extracts significantly diminished NO production and showed noteworthy reductions in the mRNA expression levels of the aforementioned genes. Furthermore, while Nrf2 is typically associated with modulating inflammatory responses, our findings indicate that the anti-inflammatory effects of our extracts are not Nrf2-dependent. Moreover, the chemical diversity of the potent extract (B Sh F) was elucidated using Q-TOF LC-HRMS, identifying 54 compounds, some of which played vital roles in suppressing inflammation. Most notably, compounds like granisetron, fenofibrate, and umbelliprenin were found to downregulate TNF-α, IL-1β, and IL-6 through the NF-κB signaling pathway. In conclusion, "Aspergillus unguis isolate SP51-EGY", isolated from the Red Sea, Egypt, has been unveiled as a promising TLR4 inhibitor with significant anti-inflammatory potentials, presenting novel insights for their potential therapeutic use in inflammation.
Collapse
Affiliation(s)
- Soad Nasr
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo (AUC), P.O. Box: 74, Cairo, 11835, Egypt
- Biochemical Engineering Department, Faculty of Energy and Environmental Engineering, The British University in Egypt, Suez Desert Road, P.O. Box: 43, El-Shorouk City, Cairo, 11837, Egypt
| | - Abdelhameed S Dawood
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo (AUC), P.O. Box: 74, Cairo, 11835, Egypt
| | - Amal Mosad Ibrahim
- Chemistry of Natural and Microbial Products Department, National Research Centre, Giza, 12622, Egypt
| | | | - Walid Fayad
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Giza, 12622, Egypt
| | - Anwar Abdelnaser
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo (AUC), P.O. Box: 74, Cairo, 11835, Egypt.
| | - Faten K Abd El-Hady
- Chemistry of Natural and Microbial Products Department, National Research Centre, Giza, 12622, Egypt
| |
Collapse
|
49
|
Nwabo Kamdje AH, Dongmo Fogang HP, Mimche PN. Role of epigenetic in cancer biology, in hematologic malignancies and in anticancer therapy. FRONTIERS IN MOLECULAR MEDICINE 2024; 4:1426454. [PMID: 39308891 PMCID: PMC11412843 DOI: 10.3389/fmmed.2024.1426454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/08/2024] [Indexed: 09/25/2024]
Abstract
Major epigenetic changes are associated with carcinogenesis, including aberrant DNA methylations and post-translational modifications of histone. Indeed evidence accumulated in recent years indicates that inactivating DNA hypermethylation preferentially targets the subset of polycomb group (PcG) genes that are regulators of developmental processes. Conversely, activating DNA hypomethylation targets oncogenic signaling pathway genes, but outcomes of both events lead in the overexpression of oncogenic signaling pathways that contribute to the stem-like state of cancer cells. On the basis of recent evidence from population-basedclinical and experimental studies, we hypothesize that factors associated with risk for developing a hematologic malignancy (HM), such as metabolic syndrome and chronic inflammation, may trigger epigenetic mechanisms to increase the transcriptional expression of oncogenes and activate oncogenic signaling pathways. Signaling pathways associated with such risk factors include but are not limited to pro-inflammatory nuclear factor κB (NF-κB) and mitogenic, growth, and survival Janus kinase (JAK) intracellular non-receptor tyrosine kinase-triggered pathways. The latter includes signaling pathways such as transducer and activator of transcription (STAT), Ras GTPases/mitogen-activated protein kinases (MAPKs)/extracellular signal-related kinases (ERKs), phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR), and β-catenin pathways. Recent findings on epigenetic mechanisms at work in the biology of cancer and in HMs and their importance in the etiology and pathogenesis of these diseases are herein summarized and discussed. Furthermore, the role of epigenetic processes in the determination of biological identity, the consequences for interindividual variability in disease clinical profile, and the potential of epigenetic drugs in HMs are also considered.
Collapse
Affiliation(s)
- Armel Hervé Nwabo Kamdje
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Biomedical Sciences, University of Garoua, Garoua, Cameroon
| | - Hervet Paulain Dongmo Fogang
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Biomedical Sciences, University of Garoua, Garoua, Cameroon
| | - Patrice N. Mimche
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
50
|
Fayed RH, Ali SE, Yassin AM, Madian K, Bawish BM. Terminalia bellirica and Andrographis paniculata dietary supplementation in mitigating heat stress-induced behavioral, metabolic and genetic alterations in broiler chickens. BMC Vet Res 2024; 20:388. [PMID: 39227945 PMCID: PMC11370032 DOI: 10.1186/s12917-024-04233-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/12/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Heat stress (HS) is one of the most significant environmental stressors on poultry production and welfare worldwide. Identification of innovative and effective solutions is necessary. This study evaluated the effects of phytogenic feed additives (PHY) containing Terminalia bellirica and Andrographis paniculata on behavioral patterns, hematological and biochemical parameters, Oxidative stress biomarkers, and HSP70, I-FABP2, IL10, TLR4, and mTOR genes expression in different organs of broiler chickens under chronic HS conditions. A total of 208 one-day-old Avian-480 broiler chicks were randomly allocated into four treatments (4 replicate/treatment, 52 birds/treatment): Thermoneutral control treatment (TN, fed basal diet); Thermoneutral treatment (TN, fed basal diet + 1 kg/ton feed PHY); Heat stress treatment (HS, fed basal diet); Heat stress treatment (HS, fed basal diet + 1 kg/ton feed PHY). RESULTS The findings of the study indicate that HS led to a decrease in feeding, foraging, walking, and comfort behavior while increasing drinking and resting behavior, also HS increased red, and white blood cells (RBCs and WBCs) counts, and the heterophile/ lymphocyte (H/L) ratio (P < 0.05); while both mean corpuscular volume (MCV), and mean corpuscular hemoglobin (MCH) were decreased (P < 0.05). In addition, HS negatively impacted lipid, protein, and glucose levels, liver and kidney function tests, and oxidative biomarkers by increasing malondialdehyde (MDA) levels and decreasing reduced glutathion (GSH) activity (P < 0.05). Heat stress (HS) caused the upregulation in HSP70, duodenal TLR4 gene expression, and the downregulation of I-FABP2, IL10, mTOR in all investigated tissues, and hepatic TLR4 (P < 0.05) compared with the TN treatment. Phytogenic feed additives (PHY) effectively mitigated heat stress's negative impacts on broilers via an improvement of broilers' behavior, hematological, biochemical, and oxidative stress biomarkers with a marked decrease in HSP70 expression levels while all tissues showed increased I-FABP2, IL10, TLR4, and mTOR (except liver) levels (P < 0.05). CONCLUSION Phytogenic feed additives (PHY) containing Terminalia bellirica and Andrographis paniculata have ameliorated the HS-induced oxidative stress and improved the immunity as well as the gut health and welfare of broiler chickens.
Collapse
Affiliation(s)
- Rabie H Fayed
- Department of Veterinary Hygiene and Management, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Sara E Ali
- Department of Physiology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Aya M Yassin
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
| | - K Madian
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Basma M Bawish
- Department of Veterinary Hygiene and Management, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| |
Collapse
|