1
|
Laroche VT, Cavill R, Kouhsar M, Reijnders RA, Harvey J, Smith AR, Imm J, Koetsier J, Weymouth L, MacBean L, Pegoraro G, Eijssen L, Creese B, Kenis G, Tijms BM, van den Hove D, Lunnon K, Pishva E. Epigenomic subtypes of late-onset Alzheimer's disease reveal distinct microglial signatures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.15.643144. [PMID: 40166175 PMCID: PMC11957029 DOI: 10.1101/2025.03.15.643144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Growing evidence suggests that clinical, pathological, and genetic heterogeneity in late-onset Alzheimer's disease contributes to variable therapeutic outcomes, potentially explaining many trial failures. Advances in molecular subtyping through proteomic and transcriptomic profiling reveal distinct patient subgroups, highlighting disease complexity beyond amyloid-beta plaques and tau tangles. This insight underscores the need to expand molecular subtyping across new molecular layers, to identify novel drug targets for different patient subgroups. In this study, we analyzed genome-wide DNA methylation data from three independent postmortem brain cohorts (n = 831) to identify epigenetic subtypes of late-onset Alzheimer's disease. Unsupervised clustering approaches were employed to identify distinct DNA methylation patterns, with subsequent cross-cohort validation to ensure robustness and reproducibility. To explore the cell-type specificity of the identified epigenomic subtypes, we characterized their methylation signatures utilizing DNA methylation profiles derived from purified brain cells. Transcriptomic data from bulk and single-cell RNA sequencing were integrated to examine the functional impact of epigenetic subtypes on gene expression profiles. Finally, we performed statistical analyses to investigate associations between these DNA methylation-defined subtypes and clinical or neuropathological features, aiming to elucidate their biological significance and clinical implications. We identified two distinct epigenomic subtypes of late-onset Alzheimer's disease, each defined by reproducible DNA methylation patterns across three cohorts. Both subtypes exhibit cell-type-specific DNA methylation profiles. Subtype 1 and subtype 2 show significant microglial methylation enrichment, with odds ratios (OR) of 1.6 and 1.3, respectively. The minimal overlap between them suggests distinct microglial states. Additionally, subtype 2 displays strong neuronal (OR = 1.6) and oligodendrocyte (OR = 3.6) enrichment. Bulk transcriptomic analyses further highlighted divergent biological mechanisms underpinning these subtypes, with subtype 1 enriched for immune-related processes, and subtype 2 characterized predominantly by neuronal and synaptic functional pathways. Single-cell transcriptional profiling of microglia revealed subtype-specific inflammatory states: subtype 1 represented a state of chronic innate immune hyperactivation with impaired resolution, while subtype 2 exhibited a more dynamic inflammatory profile balancing pro-inflammatory signaling with reparative and regulatory mechanisms. This study highlights the molecular heterogeneity of late-onset Alzheimer's disease by identifying two epigenetic subtypes with distinct cell-type-specific DNA methylation patterns. Their alignment with previously defined molecular classifications underscores their relevance in disease pathogenesis. By linking these subtypes to inflammatory microglial activity, our findings provide a foundation for future precision medicine approaches in Alzheimer's research and treatment.
Collapse
Affiliation(s)
- Valentin T. Laroche
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Rachel Cavill
- Department of Advanced Computing Sciences, Faculty of Science and Engineering (FSE), Maastricht University, Maastricht, The Netherlands
| | - Morteza Kouhsar
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Rick A. Reijnders
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Joshua Harvey
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Adam R. Smith
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Jennifer Imm
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Jarno Koetsier
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Luke Weymouth
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Lachlan MacBean
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Giulia Pegoraro
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Lars Eijssen
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Byron Creese
- Department of Life Sciences, Brunel University, London, UK
| | - Gunter Kenis
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Betty M. Tijms
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Daniel van den Hove
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Katie Lunnon
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Ehsan Pishva
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
2
|
Feng Y, Zhou Q, Hu B, Wang S, Chen L, Cai W, Zhu Q, Qin X, Zhou W, Wu Y, Song W. Unveiling the role of KLF9-mediated IFITM3 regulation in amyloidogenesis. FASEB J 2025; 39:e70403. [PMID: 39953787 DOI: 10.1096/fj.202401584rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/27/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Interferon-induced transmembrane protein 3 (IFITM3) is implicated in the pathogenesis of Alzheimer's Disease (AD) by regulating γ-secretase activity and subsequent amyloid β (Aβ) generation. However, the regulation of IFITM3 gene expression and the underlying mechanisms remain exclusive. In this study, we aimed to investigate the regulation of the IFITM3 and its role in amyloidogenesis. The functional active promoter of the IFITM3 gene was identified within the 1047 bp of 5'-flanking regions by luciferase assays. Through chromatin immunoprecipitation (ChIP) and electrophoretic mobility shift assay (EMSA), we successfully identified a specific Krüppel-like factor 9 (KLF9) binding site within the promoter region. Moreover, KLF9 overexpression significantly upregulates IFITM3 expression in vitro and in vivo, which promotes Aβ generation in the hippocampus of mice. Consistently, reduced IFITM3 expression results in a notable decrease of Aβ production. Together, we demonstrate that KLF9 plays a critical role in regulating IFITM3 expression and subsequent Aβ production. It highly suggests that inhibiting KLF9-mediated IFITM3 expression may have therapeutic potential for AD by reducing Aβ production.
Collapse
Affiliation(s)
- Yijia Feng
- Center for Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key Laboratory of Basic and Translational Research for Mental Disorders, Zhejiang Provincial Clinical Research Center for Mental Disorders, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and the Affiliated Wenzhou Kangning Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China
| | - Qian Zhou
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Bolang Hu
- Center for Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key Laboratory of Basic and Translational Research for Mental Disorders, Zhejiang Provincial Clinical Research Center for Mental Disorders, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and the Affiliated Wenzhou Kangning Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China
| | - Shengya Wang
- Center for Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key Laboratory of Basic and Translational Research for Mental Disorders, Zhejiang Provincial Clinical Research Center for Mental Disorders, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and the Affiliated Wenzhou Kangning Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lifen Chen
- Center for Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key Laboratory of Basic and Translational Research for Mental Disorders, Zhejiang Provincial Clinical Research Center for Mental Disorders, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and the Affiliated Wenzhou Kangning Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wantong Cai
- Center for Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China
| | - Qinxin Zhu
- Center for Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key Laboratory of Basic and Translational Research for Mental Disorders, Zhejiang Provincial Clinical Research Center for Mental Disorders, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and the Affiliated Wenzhou Kangning Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuemei Qin
- Center for Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key Laboratory of Basic and Translational Research for Mental Disorders, Zhejiang Provincial Clinical Research Center for Mental Disorders, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and the Affiliated Wenzhou Kangning Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weihui Zhou
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yili Wu
- Wenzhou Key Laboratory of Basic and Translational Research for Mental Disorders, Zhejiang Provincial Clinical Research Center for Mental Disorders, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and the Affiliated Wenzhou Kangning Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China
| | - Weihong Song
- Center for Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key Laboratory of Basic and Translational Research for Mental Disorders, Zhejiang Provincial Clinical Research Center for Mental Disorders, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and the Affiliated Wenzhou Kangning Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Xie Z, Li L, Hou W, Fan Z, Zeng L, He L, Ji Y, Zhang J, Wang F, Xing Z, Wang Y, Ye Y. Critical role of Oas1g and STAT1 pathways in neuroinflammation: insights for Alzheimer's disease therapeutics. J Transl Med 2025; 23:182. [PMID: 39953505 PMCID: PMC11829366 DOI: 10.1186/s12967-025-06112-2] [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: 01/08/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) has a significant impact on an individual's health and places a heavy burden on society. Studies have emphasized the importance of microglia in the progression and development of AD. Interferon responses and Interferon-stimulated genes (ISGs) significantly function in neuroinflammatory and neurodegenerative diseases involving AD. Therefore, further exploration of the relationship among microglia, ISGs, and neuroinflammation in AD is warranted. METHODS Microglia datasets from the GEO database were retrieved, along with additional microglia RNA-seq data from laboratory mice. Weighted Correlation Network Analysis was used on the training dataset to identify gene co-expression networks. Genes from the black module were intersected with interferon-stimulated genes, and differentially expressed genes (DEGs) were identified. Machine learning algorithms were applied to DEGs, and genes selected by both methods were identified as hub genes, with ROC curves used to evaluate their diagnostic accuracy. Gene Set Enrichment Analysis was performed to reveal functional pathways closely relating to hub genes. Microglia cells were transfected with siRNAs targeting Oas1g and STAT1. Total RNA from microglia cells and mouse brain tissues was extracted, reverse-transcribed, and analyzed via qRT-PCR. Proteins were extracted from cells, quantified, separated by SDS-PAGE, transferred to PVDF membranes, and probed with antibodies. Microglia cells were fixed, permeabilized, blocked, and stained with antibodies for STAT1, then visualized and photographed. RESULTS Bioinformatics and machine learning algorithms revealed that Oas1g was identified as a hub gene, with an AUC of 0.812. Enrichment Analysis revealed that Oas1g is closely associated with interferon-related pathways. Expression of Oas1g was validated in AD mouse models, where it was significantly upregulated after microglial activation. Knockdown experiments suggested siOas1g attenuated the effect of siSTAT1, and the expressions of STAT1 and p-STAT1 were elevated. siOas1g could reverse the effect of siSTAT1, indicating that Oas1g potentially regulates the ISGs through the STAT1 pathway. CONCLUSION We demonstrated that Oas1g was identified as a hub ISG in AD and can downregulate the activation of IFN-β and STAT1, reducing the expression of ISGs in neuroinflammation. Oas1g might potentially be a beneficial candidate for both prevention and treatment of AD.
Collapse
Affiliation(s)
- Zhixin Xie
- The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou, China
| | - Linxi Li
- Department of Neurosurgery, Institute of Neuroscience, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Weizhong Hou
- The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou, China
| | - Zhongxi Fan
- The Third Clinical Medicine School, Guangzhou Medical University, Guangzhou, China
| | - Lifan Zeng
- The Third Clinical Medicine School, Guangzhou Medical University, Guangzhou, China
| | - Limin He
- The Sixth Clinical Medicine School, Guangzhou Medical University, Guangzhou, China
| | - Yunxiang Ji
- Department of Neurosurgery, Institute of Neuroscience, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jingbai Zhang
- Department of Neurosurgery, Institute of Neuroscience, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fangran Wang
- Department of Neurosurgery, Institute of Neuroscience, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhou Xing
- Department of Neurosurgery, Institute of Neuroscience, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Yezhong Wang
- Department of Neurosurgery, Institute of Neuroscience, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Yongyi Ye
- Department of Neurosurgery, Institute of Neuroscience, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
4
|
Feng Y, Wang S, Yang D, Zheng W, Xia H, Zhu Q, Wang Z, Hu B, Jiang X, Qin X, Ni C, Pan W, Zhao Y, Pan S, Zhang Y, Song W. Inhibition of IFITM3 in cerebrovascular endothelium alleviates Alzheimer's-related phenotypes. Alzheimers Dement 2025; 21:e14543. [PMID: 39807629 PMCID: PMC11851164 DOI: 10.1002/alz.14543] [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: 08/30/2024] [Revised: 11/26/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025]
Abstract
INTRODUCTION Interferon-induced transmembrane protein 3 (IFITM3) modulates γ-secretase in Alzheimer's Disease (AD). Although IFITM3 knockout reduces amyloid β protein (Aβ) production, its cell-specific effect on AD remains unclear. METHODS Single nucleus RNA sequencing (snRNA-seq) was used to assess IFITM3 expression. Adeno-associated virus-BI30 (AAV-BI30) was injected to reduce IFITM3 expression in the cerebrovascular endothelial cells (CVECs). The effects on AD phenotypes in cells and AD mice were examined through behavioral tests, two-photon imaging, flow cytometry, Western blot, immunohistochemistry, and quantitative polymerase chain reaction assay (qPCR). RESULTS IFITM3 expression was increased in the CVECs of patients with AD. Overexpression of IFITM3 in primary endothelial cells enhanced Aβ generation through regulating beta-site APP cleaving enzyme 1 (BACE1) and γ-secretase. Aβ further increased IFITM3 expression, creating a vicious cycle. Knockdown of IFITM3 in CVECs decreased Aβ accumulation within cerebrovascular walls, reduced Alzheimer's-related pathology, and improved cognitive performance in AD transgenic mice. DISCUSSION Knockdown of IFITM3 in CVECs alleviates AD pathology and cognitive impairment. Targeting cerebrovascular endothelial IFITM3 holds promise for AD treatment. HIGHLIGHTS Interferon-induced transmembrane protein 3 (IFITM3) expression was increased in the cerebrovascular endothelial cells (CVECs) of patients with Alzheimer's Disease (AD). Cerebrovascular endothelial IFITM3 regulates amyloid β protein (Aβ) generation through regulating beta-site APP cleaving enzyme 1 (BACE1) and γ-secretase. Knockdown of IFITM3 in CVECs reduces Aβ deposits and improves cognitive impairments in AD transgenic mice. Cerebrovascular endothelial IFITM3 could be a potential target for the treatment of AD.
Collapse
Affiliation(s)
- Yijia Feng
- Center for Geriatric MedicineKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceThe First Affiliated Hospital and Institute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Shengya Wang
- Center for Geriatric MedicineKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceThe First Affiliated Hospital and Institute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Danlu Yang
- Center for Geriatric MedicineKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceThe First Affiliated Hospital and Institute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Wu Zheng
- Neuroscience Medical CenterNingbo Medical Center Lihuili HospitalNingbo UniversityNingboZhejiangChina
| | - Huwei Xia
- Center for Geriatric MedicineKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceThe First Affiliated Hospital and Institute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Qinxin Zhu
- Center for Geriatric MedicineKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceThe First Affiliated Hospital and Institute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Zhipeng Wang
- Center for Geriatric MedicineKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceThe First Affiliated Hospital and Institute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Bolang Hu
- Center for Geriatric MedicineKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceThe First Affiliated Hospital and Institute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Xinyi Jiang
- Center for Geriatric MedicineKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceThe First Affiliated Hospital and Institute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Xuemei Qin
- Center for Geriatric MedicineKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceThe First Affiliated Hospital and Institute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Chenkang Ni
- Center for Geriatric MedicineKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceThe First Affiliated Hospital and Institute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Wenhao Pan
- Center for Geriatric MedicineKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceThe First Affiliated Hospital and Institute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Yifan Zhao
- Center for Geriatric MedicineKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceThe First Affiliated Hospital and Institute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
| | - Sipei Pan
- Center for Geriatric MedicineKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceThe First Affiliated Hospital and Institute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| | - Yun Zhang
- Department of NeurologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Weihong Song
- Center for Geriatric MedicineKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceThe First Affiliated Hospital and Institute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| |
Collapse
|
5
|
Arachchi UPE, Madushani KP, Shanaka KASN, Kim G, Lim C, Yang H, Jayamali BPMV, Kodagoda YK, Warnakula WADLR, Jung S, Wan Q, Lee J. Characterization of tripartite motif containing 59 (TRIM59) in Epinephelus akaara: Insights into its immune involvement and functional properties in viral pathogenesis, macrophage polarization, and apoptosis regulation. FISH & SHELLFISH IMMUNOLOGY 2025; 157:110082. [PMID: 39645217 DOI: 10.1016/j.fsi.2024.110082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/21/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
The tripartite motif-containing (TRIM) superfamily is the largest family of RING-type E3 ubiquitin ligases that is conserved across the metazoan kingdom. Previous studies in mammals have demonstrated that TRIM59 possesses ubiquitin-protein ligase activity and acts as a negative regulator of NF-κB signaling. However, TRIM59 has rarely been characterized in fish. This study aimed to characterize TRIM59 from Epinephelus akaara (Eatrim59) and elucidate its structural features, expression patterns, and functional properties in innate immune responses and in the regulation of apoptosis. Eatrim59 is composed of 406 amino acids with a molecular weight of 45.84 kDa and a theoretical isoelectric point of 5.25. It comprises a conserved RING domain, a B-box motif, and a coiled-coil region. Subcellular localization analysis revealed that Eatrim59 was localized in the endoplasmic reticulum. Eatrim59 was ubiquitously expressed in all tissues examined, with the highest relative expression detected in the blood, followed by the brain and spleen. Temporal expression of Eatrim59 was dynamically regulated in response to in vivo immune stimulation by Toll-like receptor ligands and nervous necrosis virus infection. In FHM cells overexpressing Eatrim59, an increase in viral replication was observed upon infection with the Viral hemorrhagic septicemia virus. This phenomenon is attributed to Eatrim59-mediated downregulation of interferon, pro-inflammatory cytokines, and other antiviral pathways. Moreover, macrophages stably overexpressing Eatrim59 exhibited a decrease in nitric oxide production and the formation of a filamentous actin structure upon lipopolysaccharide stimulation, indicating dampened M1 polarization. Furthermore, a decrease in apoptosis was observed in Eatrim59-overexpressing FHM cells under oxidative stress induced by H2O2. In conclusion, these findings demonstrate the multifaceted role of Eatrim59 as a regulator of innate immune response and apoptosis in E. akaara.
Collapse
Affiliation(s)
- U P E Arachchi
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - K P Madushani
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - K A S N Shanaka
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Gaeun Kim
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Chaehyeon Lim
- Genetics and Breeding Research Center, National Institute of Fisheries Science, Geoje, 53334, Republic of Korea
| | - Hyerim Yang
- Genetics and Breeding Research Center, National Institute of Fisheries Science, Geoje, 53334, Republic of Korea
| | - B P M Vileka Jayamali
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Yasara Kavindi Kodagoda
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - W A D L R Warnakula
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Sumi Jung
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Jeju National University, Jeju, 63333, Republic of Korea
| | - Qiang Wan
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Jeju National University, Jeju, 63333, Republic of Korea
| | - Jehee Lee
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Jeju National University, Jeju, 63333, Republic of Korea.
| |
Collapse
|
6
|
Yang M, Su Y, Xu K, Wen P, Xie J, Wan X, Jing W, Yang Z, Liu L, Xu P. Viral infections of the central nervous system increase the risk of knee osteoarthritis: a two-sample mendelian randomization study. Aging Clin Exp Res 2025; 37:30. [PMID: 39836329 PMCID: PMC11750930 DOI: 10.1007/s40520-025-02927-7] [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: 08/20/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVE Osteoarthritis (OA) represents a condition under the influence of central nervous system (CNS) regulatory mechanisms. This investigation aims to examine the causal association between viral infections of the central nervous system (VICNS) and inflammatory diseases of the central nervous system (IDCNS) and knee osteoarthritis (KOA) at the genetic level. METHODS In this investigation, VICNS and IDCNS were considered as primary exposure variables, while KOA served as the primary outcome. Employing a two-sample mendelian randomization (MR) approach, we conducted an analysis utilizing summary data derived from genome-wide association studies (GWAS). The GWAS summary data pertaining to VICNS and IDCNS were procured from the Finnish consortium, whereas the IEU OpenGWAS database furnished the requisite data for KOA. To ensure the robustness of our genetic causal assessment, a comprehensive array of sensitivity analyses was undertaken, encompassing evaluations of heterogeneity, horizontal pleiotropy, outlier identification, leave-one-out analyses, and assessment of the normal distribution. RESULTS The results of the MR analyses revealed a suggestive positive genetic causal relationship between VICNS and KOA (P = 0.012, odds ratio [OR] with a 95% confidence interval [CI] of 1.033 [1.007-1.059]). Conversely, the MR analyses did not indicate any evidence of genetic causation between IDCNS and KOA (P = 0.575, OR 95% CI = 0.986 [0.940-1.035]). Importantly, the genetic causal assessment of the exposure and outcome variables did not demonstrate any indications of heterogeneity, horizontal pleiotropy, or outliers. Furthermore, this assessment remained robust against the influence of individual single nucleotide polymorphisms (SNPs) and exhibited adherence to a normal distribution. CONCLUSION The result of this study has elucidated a suggestive positive genetic causal link between the VICNS and KOA. However, no such genetic causal relationship was observed between the IDCNS and KOA. These findings substantiate the genetic underpinnings supporting the association between the CNS and OA.
Collapse
Affiliation(s)
- Mingyi Yang
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an, Shaanxi, 710054, China
| | - Yani Su
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China
| | - Ke Xu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an, Shaanxi, 710054, China
| | - Pengfei Wen
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China
| | - Jiale Xie
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an, Shaanxi, 710054, China
| | - Xianjie Wan
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an, Shaanxi, 710054, China
| | - Wensen Jing
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China
| | - Zhi Yang
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China
| | - Lin Liu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China.
| | - Peng Xu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China.
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an, Shaanxi, 710054, China.
| |
Collapse
|
7
|
Tagliatti E, Bizzotto M, Morini R, Filipello F, Rasile M, Matteoli M. Prenatal drivers of microglia vulnerability in the adult. Immunol Rev 2024; 327:100-110. [PMID: 39508795 DOI: 10.1111/imr.13418] [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: 11/15/2024]
Abstract
Environmental insults during early development heavily affect brain trajectories. Among these, maternal infections, high-fat diet regimens, and sleep disturbances pose a significant risk for neurodevelopmental derangements in the offspring. Notably, scattered evidence is starting to emerge that also paternal lifestyle habits may impact the offspring development. Given their key role in controlling neurogenesis, synaptogenesis and shaping neuronal circuits, microglia represent the most likely suspects of mediating the detrimental effects of prenatal insults. For some of these environmental triggers, like maternal infections, ample literature evidence demonstrates the central role of microglia, also delineating the specific transcriptomic and proteomic profiles induced by these insults. In other contexts, the analysis of microglia is still in its infancy. Fostering these studies is needed to define microglia as potential therapeutic target in the frame of disorders consequent to maternal immune activation.
Collapse
Affiliation(s)
| | | | | | | | - Marco Rasile
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Michela Matteoli
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| |
Collapse
|
8
|
Xie Q, Wang L, Liao X, Huang B, Luo C, Liao G, Yuan L, Liu X, Luo H, Shu Y. Research Progress into the Biological Functions of IFITM3. Viruses 2024; 16:1543. [PMID: 39459876 PMCID: PMC11512382 DOI: 10.3390/v16101543] [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: 08/15/2024] [Revised: 09/22/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Interferon-induced transmembrane proteins (IFITMs) are upregulated by interferons. They are not only highly conserved in evolution but also structurally consistent and have almost identical structural domains and functional domains. They are all transmembrane proteins and have multiple heritable variations in genes. The IFITM protein family is closely related to a variety of biological functions, including antiviral immunity, tumor formation, bone metabolism, cell adhesion, differentiation, and intracellular signal transduction. The progress of the research on its structure and related functions, as represented by IFITM3, is reviewed.
Collapse
Affiliation(s)
- Qian Xie
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Liangliang Wang
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China;
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of Biologicals NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102629, China
| | - Xinzhong Liao
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Bi Huang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Chuming Luo
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Guancheng Liao
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Lifang Yuan
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Xuejie Liu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Huanle Luo
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Yuelong Shu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
- Key Laboratory of Pathogen Infection Prevention and Control (MOE), State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 102629, China
| |
Collapse
|
9
|
Marshall EM, Bauer L, Nelemans T, Sooksawasdi Na Ayudhya S, Benavides F, Lanko K, de Vrij FMS, Kushner SA, Koopmans M, van Riel D, Rockx B. Differential susceptibility of human motor neurons to infection with Usutu and West Nile virus. J Neuroinflammation 2024; 21:236. [PMID: 39334427 PMCID: PMC11437828 DOI: 10.1186/s12974-024-03228-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
West Nile virus (WNV) and Usutu virus (USUV) are closely related flaviviruses with differing capacities to cause neurological disease in humans. WNV is thought to use a transneural route of neuroinvasion along motor neurons and causes severe motor deficits. The potential for use of transneural routes of neuroinvasion by USUV has not been investigated experimentally, and evidence from the few clinical case reports of USUV-associated neuroinvasive disease is lacking. We hypothesised that, compared with WNV, USUV is less able to infect motor neurons, and therefore determined the susceptibility of human induced pluripotent stem cell (iPSC)-derived spinal cord motor neurons to infection. Both viruses could grow to high titres in iPSC-derived neural cultures. However, USUV could not productively infect motor neurons due to restriction by the antiviral response, which was not induced upon WNV infection. Inhibition of the antiviral response allowed for widespread infection and transportation of USUV along motor neurons within a compartmented culture system. These results show a stark difference in the ability of these two viruses to evade initiation of intrinsic antiviral immunity. Our data suggests that USUV cannot infect motor neurons in healthy individuals but in case of immunodeficiency may pose a risk for motor-related neurological disease and transneural invasion.
Collapse
Affiliation(s)
- Eleanor M Marshall
- Department of Viroscience, Erasmus Medical Center, Dr. Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
| | - Lisa Bauer
- Department of Viroscience, Erasmus Medical Center, Dr. Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
| | - Tessa Nelemans
- Department of Medical Microbiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Syriam Sooksawasdi Na Ayudhya
- Department of Viroscience, Erasmus Medical Center, Dr. Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
- Faculty of Veterinary Science, Prince of Songkla University, Songkhla, Thailand
| | - Feline Benavides
- Department of Viroscience, Erasmus Medical Center, Dr. Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
| | - Kristina Lanko
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Femke M S de Vrij
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Steven A Kushner
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Marion Koopmans
- Department of Viroscience, Erasmus Medical Center, Dr. Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
| | - Debby van Riel
- Department of Viroscience, Erasmus Medical Center, Dr. Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
| | - Barry Rockx
- Department of Viroscience, Erasmus Medical Center, Dr. Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands.
| |
Collapse
|
10
|
Joshi R, Brezani V, Mey GM, Guixé-Muntet S, Ortega-Ribera M, Zhuang Y, Zivny A, Werneburg S, Gracia-Sancho J, Szabo G. IRF3 regulates neuroinflammatory responses and the expression of genes associated with Alzheimer's disease. J Neuroinflammation 2024; 21:212. [PMID: 39215356 PMCID: PMC11363437 DOI: 10.1186/s12974-024-03203-7] [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: 03/11/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
The pathological role of interferon signaling is emerging in neuroinflammatory disorders, yet, the specific role of Interferon Regulatory Factor 3 (IRF3) in neuroinflammation remains poorly understood. Here, we show that global IRF3 deficiency delays TLR4-mediated signaling in microglia and attenuates the hallmark features of LPS-induced inflammation such as cytokine release, microglial reactivity, astrocyte activation, myeloid cell infiltration, and inflammasome activation. Moreover, expression of a constitutively active IRF3 (S388D/S390D: IRF3-2D) in microglia induces a transcriptional program reminiscent of the Activated Response Microglia and the expression of genes associated with Alzheimer's disease, notably apolipoprotein-e. Using bulk-RNAseq of IRF3-2D brain myeloid cells, we identified Z-DNA binding protein-1 (ZBP1) as a target of IRF3 that is relevant across various neuroinflammatory disorders. Lastly, we show IRF3 phosphorylation and IRF3-dependent ZBP1 induction in response to Aβ in primary microglia cultures. Together, our results identify IRF3 as an important regulator of LPS and Aβ -mediated neuroinflammatory responses and highlight IRF3 as a central regulator of disease-specific gene activation in different neuroinflammatory diseases.
Collapse
Affiliation(s)
- Radhika Joshi
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| | - Veronika Brezani
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| | - Gabrielle M Mey
- Department of Opthalmology and Visual Sciences, Kellogg Eye Center Michigan Neuroscience Institute, University of Michigan, Ann Arbor, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Sergi Guixé-Muntet
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute-CIBEREHD, Barcelona, Spain
| | - Marti Ortega-Ribera
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| | - Yuan Zhuang
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| | - Adam Zivny
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| | - Sebastian Werneburg
- Department of Opthalmology and Visual Sciences, Kellogg Eye Center Michigan Neuroscience Institute, University of Michigan, Ann Arbor, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jordi Gracia-Sancho
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute-CIBEREHD, Barcelona, Spain
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Gyongyi Szabo
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA.
| |
Collapse
|
11
|
Maurya SK, Borgonovo JE, Biswal S, Martínez-Cerdeño V, Mishra R, Muñoz EM. Editorial: Trends in neuroimmunology: cross-talk between brain-resident and peripheral immune cells in both health and disease. Front Immunol 2024; 15:1442322. [PMID: 39026666 PMCID: PMC11256089 DOI: 10.3389/fimmu.2024.1442322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 06/11/2024] [Indexed: 07/20/2024] Open
Affiliation(s)
- Shashank K. Maurya
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Janina E. Borgonovo
- Integrative Biology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Suryanarayan Biswal
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Verónica Martínez-Cerdeño
- Department of Pathology and Laboratory Medicine, Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children of Northern California, and MIND Institute at the UC Davis Medical Center, University of California, Davis School of Medicine, Sacramento, CA, United States
| | - Rajnikant Mishra
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Estela M. Muñoz
- Institute of Histology and Embryology of Mendoza (IHEM), National University of Cuyo (UNCuyo), National Scientific and Technical Research Council (CONICET), Mendoza, Argentina
| |
Collapse
|
12
|
Saleh RO, Jasim SA, Kadhum WR, Hjazi A, Faraz A, Abid MK, Yumashev A, Alawadi A, Aiad IAZ, Alsalamy A. Exploring the detailed role of interleukins in cancer: A comprehensive review of literature. Pathol Res Pract 2024; 257:155284. [PMID: 38663179 DOI: 10.1016/j.prp.2024.155284] [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: 02/06/2024] [Revised: 03/25/2024] [Accepted: 03/30/2024] [Indexed: 05/12/2024]
Abstract
The cancer cells that are not normal can grow into tumors, invade surrounding tissues, and travel to other parts of the body via the lymphatic or circulatory systems. Interleukins, a vital class of signaling proteins, facilitate cell-to-cell contact within the immune system. A type of non-coding RNA known as lncRNAs mediates its actions by regulating miRNA-mRNA roles (Interleukins). Because of their dual function in controlling the growth of tumors and altering the immune system's response to cancer cells, interleukins have been extensively studied concerning cancer. Understanding the complex relationships between interleukins, the immune system, the tumor microenvironment, and the components of interleukin signaling pathways that impact the miRNA-mRNA axis, including lncRNAs, has advanced significantly in cancer research. Due to the significant and all-encompassing influence of interleukins on the immune system and the development and advancement of cancers, lncRNAs play a crucial role in cancer research by modulating interleukins. Their diverse effects on immune system regulation, tumor growth encouragement, and tumor inhibition make them appealing candidates for potential cancer treatments and diagnostics. A deeper understanding of the relationship between the biology of interleukin and lncRNAs will likely result in more effective immunotherapy strategies and individualized cancer treatments.
Collapse
Affiliation(s)
- Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | - Saade Abdalkareem Jasim
- Pharmacy Department, Al-Huda University College, Anbar, Iraq; Biotechnology Department, College of Applied Science, Fallujah University, Anbar, Iraq.
| | - Wesam R Kadhum
- Department of Pharmacy, Kut University College, Kut, Wasit 52001, Iraq; Advanced Research Center, Kut University College, Kut, Wasit 52001, Iraq
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Ali Faraz
- Department of Basic Medical Sciences, College of Medicine, Majmaah University, Majmaah 11952, Saudi Arabia.
| | - Mohammed Kadhem Abid
- Department of Anesthesia, College of Health & Medical Technology, Al-Ayen University, Thi-Qar, Iraq
| | - Alexey Yumashev
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ahmed Alawadi
- College of Technical Engineering, the Islamic University, Najaf, Iraq; College of Technical Engineering, the Islamic University of Al Diwaniyah, Iraq; College of Technical Engineering, the Islamic University of Babylon, Iraq
| | - Ibrahim Ahmed Zaki Aiad
- Department of Pediatrics, General Medicine Practice Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia
| | - Ali Alsalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna 66002, Iraq
| |
Collapse
|
13
|
Staurenghi E, Testa G, Leoni V, Cecci R, Floro L, Giannelli S, Barone E, Perluigi M, Leonarduzzi G, Sottero B, Gamba P. Altered Brain Cholesterol Machinery in a Down Syndrome Mouse Model: A Possible Common Feature with Alzheimer's Disease. Antioxidants (Basel) 2024; 13:435. [PMID: 38671883 PMCID: PMC11047305 DOI: 10.3390/antiox13040435] [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: 03/06/2024] [Revised: 03/29/2024] [Accepted: 03/30/2024] [Indexed: 04/28/2024] Open
Abstract
Down syndrome (DS) is a complex chromosomal disorder considered as a genetically determined form of Alzheimer's disease (AD). Maintenance of brain cholesterol homeostasis is essential for brain functioning and development, and its dysregulation is associated with AD neuroinflammation and oxidative damage. Brain cholesterol imbalances also likely occur in DS, concurring with the precocious AD-like neurodegeneration. In this pilot study, we analyzed, in the brain of the Ts2Cje (Ts2) mouse model of DS, the expression of genes encoding key enzymes involved in cholesterol metabolism and of the levels of cholesterol and its main precursors and products of its metabolism (i.e., oxysterols). The results showed, in Ts2 mice compared to euploid mice, the downregulation of the transcription of the genes encoding the enzymes 3-hydroxy-3-methylglutaryl-CoA reductase and 24-dehydrocholesterol reductase, the latter originally recognized as an indicator of AD, and the consequent reduction in total cholesterol levels. Moreover, the expression of genes encoding enzymes responsible for brain cholesterol oxidation and the amounts of the resulting oxysterols were modified in Ts2 mouse brains, and the levels of cholesterol autoxidation products were increased, suggesting an exacerbation of cerebral oxidative stress. We also observed an enhanced inflammatory response in Ts2 mice, underlined by the upregulation of the transcription of the genes encoding for α-interferon and interleukin-6, two cytokines whose synthesis is increased in the brains of AD patients. Overall, these results suggest that DS and AD brains share cholesterol cycle derangements and altered oxysterol levels, which may contribute to the oxidative and inflammatory events involved in both diseases.
Collapse
Affiliation(s)
- Erica Staurenghi
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Valerio Leoni
- Laboratory of Clinical Pathology, Hospital Pio XI of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano-Bicocca, 20832 Desio, Italy;
| | - Rebecca Cecci
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Lucrezia Floro
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Serena Giannelli
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Eugenio Barone
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University, 00185 Roma, Italy; (E.B.); (M.P.)
| | - Marzia Perluigi
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University, 00185 Roma, Italy; (E.B.); (M.P.)
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Barbara Sottero
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Paola Gamba
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| |
Collapse
|
14
|
Costa B, Vale N. Virus-Induced Epilepsy vs. Epilepsy Patients Acquiring Viral Infection: Unravelling the Complex Relationship for Precision Treatment. Int J Mol Sci 2024; 25:3730. [PMID: 38612542 PMCID: PMC11011490 DOI: 10.3390/ijms25073730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/04/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
The intricate relationship between viruses and epilepsy involves a bidirectional interaction. Certain viruses can induce epilepsy by infecting the brain, leading to inflammation, damage, or abnormal electrical activity. Conversely, epilepsy patients may be more susceptible to viral infections due to factors, such as compromised immune systems, anticonvulsant drugs, or surgical interventions. Neuroinflammation, a common factor in both scenarios, exhibits onset, duration, intensity, and consequence variations. It can modulate epileptogenesis, increase seizure susceptibility, and impact anticonvulsant drug pharmacokinetics, immune system function, and brain physiology. Viral infections significantly impact the clinical management of epilepsy patients, necessitating a multidisciplinary approach encompassing diagnosis, prevention, and treatment of both conditions. We delved into the dual dynamics of viruses inducing epilepsy and epilepsy patients acquiring viruses, examining the unique features of each case. For virus-induced epilepsy, we specify virus types, elucidate mechanisms of epilepsy induction, emphasize neuroinflammation's impact, and analyze its effects on anticonvulsant drug pharmacokinetics. Conversely, in epilepsy patients acquiring viruses, we detail the acquired virus, its interaction with existing epilepsy, neuroinflammation effects, and changes in anticonvulsant drug pharmacokinetics. Understanding this interplay advances precision therapies for epilepsy during viral infections, providing mechanistic insights, identifying biomarkers and therapeutic targets, and supporting optimized dosing regimens. However, further studies are crucial to validate tools, discover new biomarkers and therapeutic targets, and evaluate targeted therapy safety and efficacy in diverse epilepsy and viral infection scenarios.
Collapse
Affiliation(s)
- Bárbara Costa
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal
| | - Nuno Vale
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal
| |
Collapse
|
15
|
Capendale PE, García-Rodríguez I, Ambikan AT, Mulder LA, Depla JA, Freeze E, Koen G, Calitz C, Sood V, Vieira de Sá R, Neogi U, Pajkrt D, Sridhar A, Wolthers KC. Parechovirus infection in human brain organoids: host innate inflammatory response and not neuro-infectivity correlates to neurologic disease. Nat Commun 2024; 15:2532. [PMID: 38514653 PMCID: PMC10958052 DOI: 10.1038/s41467-024-46634-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 02/29/2024] [Indexed: 03/23/2024] Open
Abstract
Picornaviruses are a leading cause of central nervous system (CNS) infections. While genotypes such as parechovirus A3 (PeV-A3) and echovirus 11 (E11) can elicit severe neurological disease, the highly prevalent PeV-A1 is not associated with CNS disease. Here, we expand our current understanding of these differences in PeV-A CNS disease using human brain organoids and clinical isolates of the two PeV-A genotypes. Our data indicate that PeV-A1 and A3 specific differences in neurological disease are not due to infectivity of CNS cells as both viruses productively infect brain organoids with a similar cell tropism. Proteomic analysis shows that PeV-A infection significantly alters the host cell metabolism. The inflammatory response following PeV-A3 (and E11 infection) is significantly more potent than that upon PeV-A1 infection. Collectively, our findings align with clinical observations and suggest a role for neuroinflammation, rather than viral replication, in PeV-A3 (and E11) infection.
Collapse
Affiliation(s)
- Pamela E Capendale
- OrganoVIR Labs, Emma Children's Hospital, Department of Pediatric Infectious Diseases, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, Amsterdam Institute for Reproduction and Development, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Inés García-Rodríguez
- OrganoVIR Labs, Emma Children's Hospital, Department of Pediatric Infectious Diseases, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, Amsterdam Institute for Reproduction and Development, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Anoop T Ambikan
- The Systems Virology Lab, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Campus Flemingsberg, Stockholm, Sweden
| | - Lance A Mulder
- OrganoVIR Labs, Emma Children's Hospital, Department of Pediatric Infectious Diseases, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, Amsterdam Institute for Reproduction and Development, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Josse A Depla
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- UniQure Biopharma B.V., Department of Research & Development, Paasheuvelweg 25A, Amsterdam, The Netherlands
| | - Eline Freeze
- OrganoVIR Labs, Emma Children's Hospital, Department of Pediatric Infectious Diseases, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, Amsterdam Institute for Reproduction and Development, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Gerrit Koen
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Carlemi Calitz
- OrganoVIR Labs, Emma Children's Hospital, Department of Pediatric Infectious Diseases, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, Amsterdam Institute for Reproduction and Development, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Vikas Sood
- The Systems Virology Lab, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Campus Flemingsberg, Stockholm, Sweden
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Renata Vieira de Sá
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- The Systems Virology Lab, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Campus Flemingsberg, Stockholm, Sweden
| | - Ujjwal Neogi
- The Systems Virology Lab, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Campus Flemingsberg, Stockholm, Sweden
| | - Dasja Pajkrt
- OrganoVIR Labs, Emma Children's Hospital, Department of Pediatric Infectious Diseases, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, Amsterdam Institute for Reproduction and Development, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Adithya Sridhar
- OrganoVIR Labs, Emma Children's Hospital, Department of Pediatric Infectious Diseases, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, Amsterdam Institute for Reproduction and Development, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, The Netherlands
| | - Katja C Wolthers
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC, Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.
| |
Collapse
|
16
|
Gu J, Zhang J, Liu Q, Xu S. Neurological risks of COVID-19 in women: the complex immunology underpinning sex differences. Front Immunol 2023; 14:1281310. [PMID: 38035090 PMCID: PMC10685449 DOI: 10.3389/fimmu.2023.1281310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
The COVID-19 pandemic has uncovered many mysteries about SARS-CoV-2, including its potential to trigger abnormal autoimmune responses. Emerging evidence suggests women may face higher risks from COVID-induced autoimmunity manifesting as persistent neurological symptoms. Elucidating the mechanisms underlying this female susceptibility is now imperative. We synthesize key insights from existing studies on how COVID-19 infection can lead to immune tolerance loss, enabling autoreactive antibodies and lymphocyte production. These antibodies and lymphocytes infiltrate the central nervous system. Female sex hormones like estrogen and X-chromosome mediated effects likely contribute to dysregulated humoral immunity and cytokine profiles among women, increasing their predisposition. COVID-19 may also disrupt the delicate immunological balance of the female microbiome. These perturbations precipitate damage to neural damage through mechanisms like demyelination, neuroinflammation, and neurodegeneration - consistent with the observed neurological sequelae in women. An intentional focus on elucidating sex differences in COVID-19 pathogenesis is now needed to inform prognosis assessments and tailored interventions for female patients. From clinical monitoring to evaluating emerging immunomodulatory therapies, a nuanced women-centered approach considering the hormonal status and immunobiology will be vital to ensure equitable outcomes. Overall, deeper insights into the apparent female specificity of COVID-induced autoimmunity will accelerate the development of solutions mitigating associated neurological harm.
Collapse
Affiliation(s)
- Jienan Gu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiale Zhang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qianhui Liu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shijie Xu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
17
|
Pereira PDC, Diniz DG, da Costa ER, Magalhães NGDM, da Silva ADJF, Leite JGS, Almeida NIP, Cunha KDN, de Melo MAD, Vasconcelos PFDC, Diniz JAP, Brites D, Anthony DC, Diniz CWP, Guerreiro-Diniz C. Genes, inflammatory response, tolerance, and resistance to virus infections in migratory birds, bats, and rodents. Front Immunol 2023; 14:1239572. [PMID: 37711609 PMCID: PMC10497949 DOI: 10.3389/fimmu.2023.1239572] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Normally, the host immunological response to viral infection is coordinated to restore homeostasis and protect the individual from possible tissue damage. The two major approaches are adopted by the host to deal with the pathogen: resistance or tolerance. The nature of the responses often differs between species and between individuals of the same species. Resistance includes innate and adaptive immune responses to control virus replication. Disease tolerance relies on the immune response allowing the coexistence of infections in the host with minimal or no clinical signs, while maintaining sufficient viral replication for transmission. Here, we compared the virome of bats, rodents and migratory birds and the molecular mechanisms underlying symptomatic and asymptomatic disease progression. We also explore the influence of the host physiology and environmental influences on RNA virus expression and how it impacts on the whole brain transcriptome of seemingly healthy semipalmated sandpiper (Calidris pusilla) and spotted sandpiper (Actitis macularius). Three time points throughout the year were selected to understand the importance of longitudinal surveys in the characterization of the virome. We finally revisited evidence that upstream and downstream regulation of the inflammatory response is, respectively, associated with resistance and tolerance to viral infections.
Collapse
Affiliation(s)
- Patrick Douglas Corrêa Pereira
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
| | - Daniel Guerreiro Diniz
- Seção de Hepatologia, Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém, Pará, Brazil
- Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Laboratório de Investigações em Neurodegeneração e Infecção, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Emanuel Ramos da Costa
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
- Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Laboratório de Investigações em Neurodegeneração e Infecção, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Nara Gyzely de Morais Magalhães
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
| | - Anderson de Jesus Falcão da Silva
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
| | - Jéssica Gizele Sousa Leite
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
| | - Natan Ibraim Pires Almeida
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
| | - Kelle de Nazaré Cunha
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
| | - Mauro André Damasceno de Melo
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
| | - Pedro Fernando da Costa Vasconcelos
- Centro de Ciências Biológicas e da Saúde, Universidade do Estado do Pará, Belém, Pará, Brazil
- Seção de Arbovirologia e Febres Hemorrágicas, Instituto Evandro Chagas, Ananindeua, Pará, Brazil
| | - José Antonio Picanço Diniz
- Seção de Hepatologia, Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém, Pará, Brazil
| | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Daniel Clive Anthony
- Department of Pharmacology, Laboratory of Experimental Neuropathology, University of Oxford, Oxford, United Kingdom
| | - Cristovam Wanderley Picanço Diniz
- Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Laboratório de Investigações em Neurodegeneração e Infecção, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Cristovam Guerreiro-Diniz
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
| |
Collapse
|