1
|
Tan Q, Yang H, He Y, Shen X, Sun L, Du X, Lin G, Zhou N, Wang N, Zhou Q, Liu D, Xu X, Zhao L, Xie P. Borna disease virus 1 induces ferroptosis, contributing to lethal encephalitis. J Med Virol 2024; 96:e29945. [PMID: 39370874 DOI: 10.1002/jmv.29945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/03/2024] [Accepted: 09/24/2024] [Indexed: 10/08/2024]
Abstract
Borna disease virus 1 (BoDV-1) is a neurotropic RNA virus that has been linked to fatal BoDV-1 encephalitis (BVE) in humans. Ferroptosis represents a newly recognized kind of programmed cell death that marked by iron overload and lipid peroxidation. Various viral infections are closely related to ferroptosis. However, the link between BoDV-1 infection and ferroptosis, as well as its role in BVE pathogenesis, remains inadequately understood. Herein, we used primary rat cortical neurons, human microglial HMC3 cells, and Sprague‒Dawley rats as models. BoDV-1 infection induced ferroptosis, as ferroptosis characteristics were detected (iron overload, reactive oxygen species buildup, decreased antioxidant capacity, lipid peroxidation, and mitochondrial damage). Analysis via qRT-PCR and Western blot demonstrated that BoDV-1-induced ferroptosis was mediated through Nrf2/HO-1/SLC7a11/GPX4 antioxidant pathway suppression. Nrf2 downregulation was due to BoDV-1 infection promoting Nrf2 ubiquitination and degradation. Following BoDV-1-induced ferroptosis, the PTGS2/PGE2 signaling pathway was activated, and various intracellular lipid peroxidation products and damage-associated molecular patterns were released, contributing to BVE occurrence and progression. More importantly, inhibiting ferroptosis or the ubiquitin‒proteasome system effectively alleviated BVE. Collectively, these findings demonstrate the interaction between BoDV-1 infection and ferroptosis and reveal BoDV-1-induced ferroptosis as an underlying pathogenic mechanism of BVE.
Collapse
Affiliation(s)
- Qing Tan
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongli Yang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Emergency Medicine, The People's Hospital of Jianyang City, Chengdu, China
| | - Yong He
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xia Shen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Sun
- Department of Anaesthesia and Pain, The First People's Hospital of Chongqing Liangjiang New Area, Chongqing, China
| | - Xiaoyan Du
- Department of Neurology, The Yongchuan Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cerebrovascular Disease Research, Chongqing, China
| | - Gangqiang Lin
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Na Zhou
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Nishi Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qian Zhou
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Liu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyan Xu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Libo Zhao
- Department of Neurology, The Yongchuan Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cerebrovascular Disease Research, Chongqing, China
| | - Peng Xie
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Yan X, Ding JY, Zhang RJ, Zhang HQ, Kang L, Jia CY, Liu XY, Shen CL. FSTL1 Accelerates Nucleus Pulposus Cell Senescence and Intervertebral Disc Degeneration Through TLR4/NF-κB Pathway. Inflammation 2024; 47:1229-1247. [PMID: 38316670 DOI: 10.1007/s10753-024-01972-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/08/2024] [Accepted: 01/08/2024] [Indexed: 02/07/2024]
Abstract
Intervertebral disc degeneration (IVDD) is a major contributor to low back pain (LBP), and inflammatory factors play crucial roles in its pathogenesis. Follistatin-like 1 (FSTL1) has been reported to induce an inflammatory response in chondrocytes, microglia and preadipocytes, but its role in the pathogenesis of nucleus pulposus cell (NPC) degeneration remains unclear. In this study, we mainly utilized an acidosis-induced NPC degeneration model and a rabbit puncture IVDD model to investigate the role of FSTL1 in IVDD both in vitro and in vivo. We confirmed that FSTL1 expression significantly increased in nucleus pulposus (NP) tissues from IVDD patients and rabbit puncture IVDD models. The expression levels of FSTL1 were significantly increased in all three models of NPC degeneration under harsh microenvironments. In addition, recombinant human FSTL1 (rh-FSTL1) was found to upregulate the expression of p16 and p21, increase the number of senescence-associated β-galactosidase (SA-β-gal)-positive cells, induce senescence-related secretory phenotypes (SASP), and downregulate extracellular matrix (ECM) protein expressions, leading to an imbalance in ECM metabolism destructions. Conversely, silencing of FSTL1 by small interfering RNA (siRNA) ameliorated senescence of NPCs associated with inflammation in IVDD. Furthermore, Toll-like receptor 4/nuclear factor-κB (TLR4/NF-κB) pathway plays a crucial role in regulating NPC senescence through FSTL1 regulation. Inhibition of TLR4 expression partly reversed the effects of rh-FSTL1 on NPC senescence-associated inflammation. Finally, rabbit IVDD model experiments demonstrated that the specific FSTL1 siRNA markedly repressed the development of IVDD. These findings may offer a therapeutic approach for mitigating inflammation-induced senescence associated with IVDD.
Collapse
Affiliation(s)
- Xu Yan
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Jing-Yu Ding
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Ren-Jie Zhang
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Hua-Qing Zhang
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Liang Kang
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Chong-Yu Jia
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Xiao-Ying Liu
- School of Life Sciences, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Cai-Liang Shen
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China.
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China.
| |
Collapse
|
3
|
Ren F, Narita R, Rashidi AS, Fruhwürth S, Gao Z, Bak RO, Thomsen MK, Verjans GMGM, Reinert LS, Paludan SR. ER stress induces caspase-2-tBID-GSDME-dependent cell death in neurons lytically infected with herpes simplex virus type 2. EMBO J 2023; 42:e113118. [PMID: 37646198 PMCID: PMC10548179 DOI: 10.15252/embj.2022113118] [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: 11/22/2022] [Revised: 08/14/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023] Open
Abstract
Neurotropic viruses, including herpes simplex virus (HSV) types 1 and 2, have the capacity to infect neurons and can cause severe diseases. This is associated with neuronal cell death, which may contribute to morbidity or even mortality if the infection is not controlled. However, the mechanistic details of HSV-induced neuronal cell death remain enigmatic. Here, we report that lytic HSV-2 infection of human neuron-like SH-SY5Y cells and primary human and murine brain cells leads to cell death mediated by gasdermin E (GSDME). HSV-2-induced GSDME-mediated cell death occurs downstream of replication-induced endoplasmic reticulum stress driven by inositol-requiring kinase 1α (IRE1α), leading to activation of caspase-2, cleavage of the pro-apoptotic protein BH3-interacting domain death agonist (BID), and mitochondria-dependent activation of caspase-3. Finally, necrotic neurons released alarmins, which activated inflammatory responses in human iPSC-derived microglia. In conclusion, lytic HSV infection in neurons activates an ER stress-driven pathway to execute GSDME-mediated cell death and promote inflammation.
Collapse
Affiliation(s)
- Fanghui Ren
- Department of BiomedicineAarhus UniversityAarhus CDenmark
| | - Ryo Narita
- Department of BiomedicineAarhus UniversityAarhus CDenmark
| | - Ahmad S Rashidi
- Department of ViroscienceErasmus Medical CentreRotterdamThe Netherlands
| | - Stefanie Fruhwürth
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgGothenburgSweden
| | - Zongliang Gao
- Department of BiomedicineAarhus UniversityAarhus CDenmark
| | - Rasmus O Bak
- Department of BiomedicineAarhus UniversityAarhus CDenmark
| | | | | | - Line S Reinert
- Department of BiomedicineAarhus UniversityAarhus CDenmark
| | - Søren R Paludan
- Department of BiomedicineAarhus UniversityAarhus CDenmark
- Department of Rheumatology and Inflammation Research, Institute of MedicineSahlgrenska Academy, University of GothenburgGothenburgSweden
| |
Collapse
|
4
|
Li K, Ma Y, Xia X, Huang H, Li J, Wang X, Gao Y, Zhang S, Fu T, Tong Y. Possible correlated signaling pathways with chronic urate nephropathy: A review. Medicine (Baltimore) 2023; 102:e34540. [PMID: 37565908 PMCID: PMC10419604 DOI: 10.1097/md.0000000000034540] [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: 04/05/2023] [Accepted: 07/11/2023] [Indexed: 08/12/2023] Open
Abstract
Hyperuricemia nephropathy, also known as gouty nephropathy, refers to renal damage induced by hyperuricemia caused by excessive production of serum uric acid or low excretion of uric acid. the persistence of symptoms will lead to changes in renal tubular phenotype and accelerate the progress of renal fibrosis. The existence and progressive aggravation of symptoms will bring a heavy burden to patients, their families and society, affect their quality of life and reduce their well-being. With the increase of reports on hyperuricemia nephropathy, the importance of related signal pathways in the pathogenesis of hyperuricemia nephropathy is becoming more and more obvious, but most studies are limited to the upper and lower mediating relationship between 1 or 2 signal pathways. The research on the comprehensiveness of signal pathways and the breadth of crosstalk between signal pathways is limited. By synthesizing the research results of signal pathways related to hyperuricemia nephropathy in recent years, this paper will explore the specific mechanism of hyperuricemia nephropathy, and provide new ideas and methods for the treatment of hyperuricemia nephropathy based on a variety of signal pathway crosstalk and personal prospects.
Collapse
Affiliation(s)
- Kaiqing Li
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yanchun Ma
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Xue Xia
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Huili Huang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Jianing Li
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Xiaoxin Wang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yang Gao
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Shuxiang Zhang
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tong Fu
- Brandeis University, Waltham, MA
| | - Ying Tong
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
5
|
Chen P, Ban W, Wang W, You Y, Yang Z. The Devastating Effects of Sleep Deprivation on Memory: Lessons from Rodent Models. Clocks Sleep 2023; 5:276-294. [PMID: 37218868 DOI: 10.3390/clockssleep5020022] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/11/2023] [Accepted: 05/11/2023] [Indexed: 05/24/2023] Open
Abstract
In this narrative review article, we discuss the role of sleep deprivation (SD) in memory processing in rodent models. Numerous studies have examined the effects of SD on memory, with the majority showing that sleep disorders negatively affect memory. Currently, a consensus has not been established on which damage mechanism is the most appropriate. This critical issue in the neuroscience of sleep remains largely unknown. This review article aims to elucidate the mechanisms that underlie the damaging effects of SD on memory. It also proposes a scientific solution that might explain some findings. We have chosen to summarize literature that is both representative and comprehensive, as well as innovative in its approach. We examined the effects of SD on memory, including synaptic plasticity, neuritis, oxidative stress, and neurotransmitters. Results provide valuable insights into the mechanisms by which SD impairs memory function.
Collapse
Affiliation(s)
- Pinqiu Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
| | - Weikang Ban
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Wenyan Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
| | - Yuyang You
- School of Automation, Beijing Institute of Technology, Beijing 100081, China
| | - Zhihong Yang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| |
Collapse
|
6
|
Fontes-Dantas FL, Fernandes GG, Gutman EG, De Lima EV, Antonio LS, Hammerle MB, Mota-Araujo HP, Colodeti LC, Araújo SM, Froz GM, da Silva TN, Duarte LA, Salvio AL, Pires KL, Leon LA, Vasconcelos CCF, Romão L, Savio LEB, Silva JL, da Costa R, Clarke JR, Da Poian AT, Alves-Leon SV, Passos GF, Figueiredo CP. SARS-CoV-2 Spike protein induces TLR4-mediated long-term cognitive dysfunction recapitulating post-COVID-19 syndrome in mice. Cell Rep 2023; 42:112189. [PMID: 36857178 PMCID: PMC9935273 DOI: 10.1016/j.celrep.2023.112189] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 12/16/2022] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Cognitive dysfunction is often reported in patients with post-coronavirus disease 2019 (COVID-19) syndrome, but its underlying mechanisms are not completely understood. Evidence suggests that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Spike protein or its fragments are released from cells during infection, reaching different tissues, including the CNS, irrespective of the presence of the viral RNA. Here, we demonstrate that brain infusion of Spike protein in mice has a late impact on cognitive function, recapitulating post-COVID-19 syndrome. We also show that neuroinflammation and hippocampal microgliosis mediate Spike-induced memory dysfunction via complement-dependent engulfment of synapses. Genetic or pharmacological blockage of Toll-like receptor 4 (TLR4) signaling protects animals against synapse elimination and memory dysfunction induced by Spike brain infusion. Accordingly, in a cohort of 86 patients who recovered from mild COVID-19, the genotype GG TLR4-2604G>A (rs10759931) is associated with poor cognitive outcome. These results identify TLR4 as a key target to investigate the long-term cognitive dysfunction after COVID-19 infection in humans and rodents.
Collapse
Affiliation(s)
- Fabricia L. Fontes-Dantas
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil,Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Gabriel G. Fernandes
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Elisa G. Gutman
- Translational Neuroscience Laboratory (LabNet), Post-Graduate Program in Neurology, Federal University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil,Clinical Medicine Post-graduation Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Emanuelle V. De Lima
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Leticia S. Antonio
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Mariana B. Hammerle
- Clinical Medicine Post-graduation Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Hannah P. Mota-Araujo
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Lilian C. Colodeti
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Suzana M.B. Araújo
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Gabrielle M. Froz
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Talita N. da Silva
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Larissa A. Duarte
- Translational Neuroscience Laboratory (LabNet), Post-Graduate Program in Neurology, Federal University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil,Clinical Medicine Post-graduation Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Andreza L. Salvio
- Translational Neuroscience Laboratory (LabNet), Post-Graduate Program in Neurology, Federal University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil
| | - Karina L. Pires
- Neurology Department, Federal University of the State of Rio de Janeiro (UNIRIO), Rio de Janeiro, RJ, Brazil
| | - Luciane A.A. Leon
- Laboratório de Desenvolvimento Tecnológico em Virologia, IOC/FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | | | - Luciana Romão
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luiz Eduardo B. Savio
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Jerson L. Silva
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Robson da Costa
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Julia R. Clarke
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Andrea T. Da Poian
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil,Corresponding author
| | - Soniza V. Alves-Leon
- Translational Neuroscience Laboratory (LabNet), Post-Graduate Program in Neurology, Federal University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil,Division of Neurology, Hospital Clementino Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil,Corresponding author
| | - Giselle F. Passos
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil,Corresponding author
| | - Claudia P. Figueiredo
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil,Corresponding author
| |
Collapse
|
7
|
Pham PH, Tockovska T, Leacy A, Iverson M, Ricker N, Susta L. Transcriptome Analysis of Duck and Chicken Brains Infected with Aquatic Bird Bornavirus-1 (ABBV-1). Viruses 2022; 14:2211. [PMID: 36298766 PMCID: PMC9611670 DOI: 10.3390/v14102211] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/02/2022] [Accepted: 10/06/2022] [Indexed: 12/21/2022] Open
Abstract
Aquatic bird bornavirus 1 (ABBV-1) is a neurotropic virus that infects waterfowls, resulting in persistent infection. Experimental infection showed that both Muscovy ducks and chickens support persistent ABBV-1 infection in the central nervous system (CNS), up to 12 weeks post-infection (wpi), without the development of clinical disease. The aim of the present study was to describe the transcriptomic profiles in the brains of experimentally infected Muscovy ducks and chickens infected with ABBV-1 at 4 and 12 wpi. Transcribed RNA was sequenced by next-generation sequencing and analyzed by principal component analysis (PCA) and differential gene expression. The functional annotation of differentially expressed genes was evaluated by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. The PCA showed that the infected ducks sampled at both 4 and 12 wpi clustered separately from the controls, while only the samples from the chickens at 12 wpi, but not at 4 wpi, formed a separate cluster. In the ducks, more genes were differentially expressed at 4 wpi than 12 wpi, and the majority of the highly differentially expressed genes (DEG) were upregulated. On the other hand, the infected chickens had fewer DEGs at 4 wpi than at 12 wpi, and the majority of those with high numbers of DEGs were downregulated at 4 wpi and upregulated at 12 wpi. The functional annotation showed that the most enriched GO terms were immune-associated in both species; however, the terms associated with the innate immune response were predominantly enriched in the ducks, whereas the chickens had enrichment of both the innate and adaptive immune response. Immune-associated pathways were also enriched according to the KEGG pathway analysis in both species. Overall, the transcriptomic analysis of the duck and chicken brains showed that the main biological responses to ABBV-1 infection were immune-associated and corresponded with the levels of inflammation in the CNS.
Collapse
Affiliation(s)
| | | | | | | | | | - Leonardo Susta
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|