1
|
Duarte RRR, Pain O, Bendall ML, de Mulder Rougvie M, Marston JL, Selvackadunco S, Troakes C, Leung SK, Bamford RA, Mill J, O'Reilly PF, Srivastava DP, Nixon DF, Powell TR. Integrating human endogenous retroviruses into transcriptome-wide association studies highlights novel risk factors for major psychiatric conditions. Nat Commun 2024; 15:3803. [PMID: 38778015 PMCID: PMC11111684 DOI: 10.1038/s41467-024-48153-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
Human endogenous retroviruses (HERVs) are repetitive elements previously implicated in major psychiatric conditions, but their role in aetiology remains unclear. Here, we perform specialised transcriptome-wide association studies that consider HERV expression quantified to precise genomic locations, using RNA sequencing and genetic data from 792 post-mortem brain samples. In Europeans, we identify 1238 HERVs with expression regulated in cis, of which 26 represent expression signals associated with psychiatric disorders, with ten being conditionally independent from neighbouring expression signals. Of these, five are additionally significant in fine-mapping analyses and thus are considered high confidence risk HERVs. These include two HERV expression signatures specific to schizophrenia risk, one shared between schizophrenia and bipolar disorder, and one specific to major depressive disorder. No robust signatures are identified for autism spectrum conditions or attention deficit hyperactivity disorder in Europeans, or for any psychiatric trait in other ancestries, although this is likely a result of relatively limited statistical power. Ultimately, our study highlights extensive HERV expression and regulation in the adult cortex, including in association with psychiatric disorder risk, therefore providing a rationale for exploring neurological HERV expression in complex neuropsychiatric traits.
Collapse
Affiliation(s)
- Rodrigo R R Duarte
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- Division of Infectious Diseases, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| | - Oliver Pain
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Matthew L Bendall
- Division of Infectious Diseases, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | | | - Jez L Marston
- Division of Infectious Diseases, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Sashika Selvackadunco
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC London Neurodegenerative Diseases Brain Bank, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Claire Troakes
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC London Neurodegenerative Diseases Brain Bank, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Szi Kay Leung
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Rosemary A Bamford
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Jonathan Mill
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Paul F O'Reilly
- Department of Genetics and Genomic Sciences, Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Douglas F Nixon
- Division of Infectious Diseases, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Timothy R Powell
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- Division of Infectious Diseases, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
2
|
Ochoa E, Ramirez P, Gonzalez E, De Mange J, Ray WJ, Bieniek KF, Frost B. Pathogenic tau-induced transposable element-derived dsRNA drives neuroinflammation. SCIENCE ADVANCES 2023; 9:eabq5423. [PMID: 36608133 PMCID: PMC9821943 DOI: 10.1126/sciadv.abq5423] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 12/02/2022] [Indexed: 06/17/2023]
Abstract
Deposition of tau protein aggregates in the brain of affected individuals is a defining feature of "tauopathies," including Alzheimer's disease. Studies of human brain tissue and various model systems of tauopathy report that toxic forms of tau negatively affect nuclear and genomic architecture, identifying pathogenic tau-induced heterochromatin decondensation and consequent retrotransposon activation as a causal mediator of neurodegeneration. On the basis of their similarity to retroviruses, retrotransposons drive neuroinflammation via toxic intermediates, including double-stranded RNA (dsRNA). We find that dsRNA and dsRNA sensing machinery are elevated in astrocytes of postmortem brain tissue from patients with Alzheimer's disease and progressive supranuclear palsy and in brains of tau transgenic mice. Using a Drosophila model of tauopathy, we identify specific tau-induced retrotransposons that form dsRNA and find that pathogenic tau and heterochromatin decondensation causally drive dsRNA-mediated neurodegeneration and neuroinflammation. Our study suggests that pathogenic tau-induced heterochromatin decondensation and retrotransposon activation cause elevation of inflammatory, transposable element-derived dsRNA in the adult brain.
Collapse
Affiliation(s)
- Elizabeth Ochoa
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Paulino Ramirez
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Elias Gonzalez
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Jasmine De Mange
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| | - William J. Ray
- The Neurodegeneration Consortium, Therapeutics Discovery Division, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kevin F. Bieniek
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Pathology and Laboratory Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Bess Frost
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
3
|
Wu L, Li S, Li C, He B, Lv L, Wang J, Wang J, Wang W, Zhang Y. The role of regulatory T cells on the activation of astrocytes in the brain of high-fat diet mice following lead exposure. Chem Biol Interact 2022; 351:109740. [PMID: 34742682 DOI: 10.1016/j.cbi.2021.109740] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 10/11/2021] [Accepted: 11/01/2021] [Indexed: 01/02/2023]
Abstract
Lead (Pb) exposure can cause damage to the central nervous system (CNS)*. Pb can accumulate in the hippocampus, leading to learning and memory impairments. Recent studies have shown that high-fat diet (HFD) is also associated with cognitive impairment. However, there are few reports on CNS damage due to HFD and Pb exposure. We aimed to investigate the effect of Pb on cognitive functions of HFD-fed mice, focusing on the role of regulatory T (Treg) cells in astrocyte activation. C57BL/6J mice were randomly divided into control, HFD, Pb, and HFD + Pb groups. TGF-β and IL-10 secreted by Treg cells and the intracellular transcription factor Foxp3 were evaluated as a measure of Treg cell function; astrocyte activation was assessed by evaluating glial fibrillary acidic protein (GFAP) expression. The learning and memory ability was significantly lower in the HFD + Pb group than in other groups. The brain Treg cell ratio was significantly decreased and the protein levels of TGF-β, IL-10, and Foxp3 were significantly lower, whereas the protein level of GFAP was higher in the HFD + Pb group. The hippocampus of the HFD + Pb group mice showed significantly higher levels of neurotoxic reactive astrocyte markers and astrogliosis was also much higher compared to HFD and Pb groups. Furthermore, all-trans retinoic acid treatment increased the brain Treg cell ratio, reversed cognitive decline, and suppressed astrocyte activation in the HFD + Pb group mice. We concluded that HFD along with Pb exposure could aggravate the activation of astrocytes in the brain, and the brain Treg cells may be involved in inhibiting astrocyte activation in HFD-fed mice exposed to Pb.
Collapse
Affiliation(s)
- Lei Wu
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Shuang Li
- Experiment Animal Center, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Chao Li
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Bin He
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Linyi Lv
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Jia Wang
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Jierui Wang
- Rheumatology Department, Kailuan General Hospital, Tangshan, 063000, Hebei, China
| | - Weixuan Wang
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Yanshu Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China; Experiment Animal Center, North China University of Science and Technology, Tangshan, 063210, Hebei, China.
| |
Collapse
|
4
|
Lopatina OL, Komleva YK, Malinovskaya NA, Panina YA, Morgun AV, Salmina AB. CD157 and Brain Immune System in (Patho)physiological Conditions: Focus on Brain Plasticity. Front Immunol 2020; 11:585294. [PMID: 33304350 PMCID: PMC7693531 DOI: 10.3389/fimmu.2020.585294] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/12/2020] [Indexed: 12/18/2022] Open
Abstract
Ectoenzyme and receptor BST-1/CD157 has been considered as a key molecule involved in the regulation of functional activity of cells in various tissues and organs. It is commonly accepted that CD157 catalyzes NAD+ hydrolysis and acts as a component of integrin adhesion receptor complex. Such properties are important for the regulatory role of CD157 in neuronal and glial cells: in addition to recently discovered role in the regulation of emotions, motor functions, and social behavior, CD157 might serve as an important component of innate immune reactions in the central nervous system. Activation of innate immune system in the brain occurs in response to infectious agents as well as in brain injury and neurodegeneration. As an example, in microglial cells, association of CD157 with CD11b/CD18 complex drives reactive gliosis and neuroinflammation evident in brain ischemia, chronic neurodegeneration, and aging. There are various non-substrate ligands of CD157 belonging to the family of extracellular matrix proteins (fibronectin, collagen I, finbrinogen, and laminin) whose activity is required for controlling cell adhesion and migration. Therefore, CD157 could control structural and functional integrity of the blood-brain barrier and barriergenesis. On the other hand, contribution of CD157 to the regulation of brain development is rather possible since in the embryonic brain, CD157 expression is very high, whereas in the adult brain, CD157 is expressed on neural stem cells and, presumably, is involved in the neurogenesis. Besides, CD157 could mediate astrocytes' action on neural stem and progenitor cells within neurogenic niches. In this review we will summarize how CD157 may affect brain plasticity acting as a molecule at the crossroad of neurogenesis, cerebral angiogenesis, and immune regulation.
Collapse
Affiliation(s)
- Olga L. Lopatina
- Department of Biochemistry, Medical, Pharmaceutical, and Toxicological Chemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
- Laboratory for Social Brain Studies, Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
- Department of Biophysics, Siberian Federal University, Krasnoyarsk, Russia
| | - Yulia K. Komleva
- Department of Biochemistry, Medical, Pharmaceutical, and Toxicological Chemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
| | - Natalia A. Malinovskaya
- Department of Biochemistry, Medical, Pharmaceutical, and Toxicological Chemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
| | - Yulia A. Panina
- Department of Biochemistry, Medical, Pharmaceutical, and Toxicological Chemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
| | - Andrey V. Morgun
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
| | - Alla B. Salmina
- Department of Biochemistry, Medical, Pharmaceutical, and Toxicological Chemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
| |
Collapse
|
5
|
Ito N, Sasaki K, Takemoto H, Kobayashi Y, Isoda H, Odaguchi H. Emotional Impairments and Neuroinflammation are Induced in Male Mice Invulnerable to Repeated Social Defeat Stress. Neuroscience 2020; 443:148-163. [PMID: 32707290 DOI: 10.1016/j.neuroscience.2020.07.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/10/2020] [Accepted: 07/14/2020] [Indexed: 11/26/2022]
Abstract
Prolonged stress triggers neuroinflammation, which plays a significant role in the development of depression; however, stressed people do not always suffer from depression because of individual differences in stress vulnerability. Negative cognitive bias (NCB) toward pessimistic judgment often underlies depressive episodes. However, a relationship between stress vulnerability, neuroinflammation, and NCB remains elusive. In addition, an animal model with all the traits would be a powerful tool for studying the etiology of depression and its therapeutic approaches. Accordingly, this study evaluated the effect of stress vulnerability on neuroinflammation and depression-related behaviors, including NCB in males, using a modified version of repeated social defeat stress (mRSDS) paradigm, a validated animal model of psychosocial stress. Exposure to mRSDS, consisting of 5 min of social defeat by unfamiliar CD-1 aggressor mice for five consecutive days, caused NCB, which co-occurred with depressive- and anxiety-like behaviors, and neuroinflammation in male BALB/c mice. Treatment with minocycline, an antibiotic with anti-inflammatory property, blocked mRSDS-induced depressive-like behaviors and neuroinflammation, but not NCB, indicating the limited effect of an anti-inflammatory intervention. In addition, marked differences were found in neuroinflammatory profiles and hippocampal gene expression patterns between resilient and unstressed mice, as well as between susceptible and resilient mice. Therefore, mice resilient to mRSDS are indeed not intact. Our findings provide insights into the unique features of the mRSDS model in male BALB/c mice, which could be used to investigate the etiological mechanisms underlying depression as well as bridge the gap in the relationship between stress vulnerability, neuroinflammation, and NCB in males.
Collapse
Affiliation(s)
- Naoki Ito
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, Minato-ku, Tokyo 108-8642, Japan.
| | - Kazunori Sasaki
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba-shi, Ibaraki 305-8572, Japan; Interdisciplinary Research Center for Catalytic Chemistry, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba-shi, Ibaraki 305-8565, Japan; Faculty of Pure and Applied Sciences, University of Tsukuba, Tsukuba-shi, Ibaraki 305-8571, Japan
| | - Hiroaki Takemoto
- School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8642, Japan
| | - Yoshinori Kobayashi
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, Minato-ku, Tokyo 108-8642, Japan; School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8642, Japan
| | - Hiroko Isoda
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba-shi, Ibaraki 305-8572, Japan; Interdisciplinary Research Center for Catalytic Chemistry, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba-shi, Ibaraki 305-8565, Japan; Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba-shi, Ibaraki 305-8572, Japan
| | - Hiroshi Odaguchi
- Department of Clinical Research, Oriental Medicine Research Center, Kitasato University, Minato-ku, Tokyo 108-8642, Japan
| |
Collapse
|
6
|
Justina VD, Giachini FR, Priviero F, Webb RC. Double-stranded RNA and Toll-like receptor activation: a novel mechanism for blood pressure regulation. Clin Sci (Lond) 2020; 134:303-313. [PMID: 31998948 PMCID: PMC7703673 DOI: 10.1042/cs20190913] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/15/2020] [Accepted: 01/17/2020] [Indexed: 12/24/2022]
Abstract
Toll-like receptors (TLRs), such as TLR4 and 9, recognize pathogen-associated molecular pattern (PAMPs) and danger-associated molecular patterns (DAMPs) and are associated with increased blood pressure (BP). TLR3, residing in the endosomal compartment, is activated by viral double-stranded RNA (dsRNA) leading to activation of TIR receptor domain-containing adaptor inducing IFN-β (TRIF) dependent pathway. Besides foreign pathogens, the immune system responds to endogenous markers of cellular damage such as mitochondrial dsRNA (mtdsRNA). New evidence has shown a link between dsRNA and increased BP. Moreover, TLR3 activation during pregnancy was demonstrated to develop preeclampsia-like symptoms in both rats and mice. Hence, we hypothesize that the dsRNA derived from viral nucleic acids or cellular damage (mtdsRNA) will increase the inflammatory state through activation of TLR3, contributing to vascular dysfunction and increased BP. Therefore, inhibition of TLR3 could be a therapeutic target for the treatment of hypertension with potential improvement in vascular reactivity and consequently, a decrease in BP.
Collapse
Affiliation(s)
- Vanessa Dela Justina
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
- Graduate Program in Biological Sciences, Federal University of Goias, Goiânia, Brazil
| | - Fernanda R. Giachini
- Graduate Program in Biological Sciences, Federal University of Goias, Goiânia, Brazil
- RIVATREM - Red Iberoamericana de Alteraciones Vasculares en Transtornos del Embarazo
| | - Fernanda Priviero
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| | - R. Clinton Webb
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| |
Collapse
|
7
|
Syk and Hrs Regulate TLR3-Mediated Antiviral Response in Murine Astrocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6927380. [PMID: 31089414 PMCID: PMC6476135 DOI: 10.1155/2019/6927380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/22/2018] [Accepted: 01/13/2019] [Indexed: 12/02/2022]
Abstract
Toll-like receptors (TLRs) sense the presence of pathogen-associated molecular patterns. Nevertheless, the mechanisms modulating TLR-triggered innate immune responses are not yet fully understood. Complex regulatory systems exist to appropriately direct immune responses against foreign or self-nucleic acids, and a critical role of hepatocyte growth factor-regulated tyrosine kinase substrate (HRS), endosomal sorting complex required for transportation-0 (ESCRT-0) subunit, has recently been implicated in the endolysosomal transportation of TLR7 and TLR9. We investigated the involvement of Syk, Hrs, and STAM in the regulation of the TLR3 signaling pathway in a murine astrocyte cell line C8-D1A following cell stimulation with a viral dsRNA mimetic. Our data uncover a relationship between TLR3 and ESCRT-0, point out Syk as dsRNA-activated kinase, and suggest the role for Syk in mediating TLR3 signaling in murine astrocytes. We show molecular events that occur shortly after dsRNA stimulation of astrocytes and result in Syk Tyr-342 phosphorylation. Further, TLR3 undergoes proteolytic processing; the resulting TLR3 N-terminal form interacts with Hrs. The knockdown of Syk and Hrs enhances TLR3-mediated antiviral response in the form of IFN-β, IL-6, and CXCL8 secretion. Understanding the role of Syk and Hrs in TLR3 immune responses is of high importance since activation and precise execution of the TLR3 signaling pathway in the brain seem to be particularly significant in mounting an effective antiviral defense. Infection of the brain with herpes simplex type 1 virus may increase the secretion of amyloid-β by neurons and astrocytes and be a causal factor in degenerative diseases such as Alzheimer's disease. Errors in TLR3 signaling, especially related to the precise regulation of the receptor transportation and degradation, need careful observation as they may disclose foundations to identify novel or sustain known therapeutic targets.
Collapse
|
8
|
An SY, Youn GS, Kim H, Choi SY, Park J. Celastrol suppresses expression of adhesion molecules and chemokines by inhibiting JNK-STAT1/NF-κB activation in poly(I:C)-stimulated astrocytes. BMB Rep 2017; 50:25-30. [PMID: 28027722 PMCID: PMC5319661 DOI: 10.5483/bmbrep.2017.50.1.114] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Indexed: 01/05/2023] Open
Abstract
In the central nervous system, viral infection can induce inflammation by up-regulating pro-inflammatory mediators that contribute to enhanced infiltration of immune cells into the central nervous areas. Celastrol is known to exert various regulatory functions, including anti-microbial activities. In this study, we investigated the regulatory effects and the mechanisms of action of celastrol against astrocytes activated with polyinosinic-polycytidylic acid (poly(I:C)), a synthetic dsRNA, as a model of pro-inflammatory mediated responses. Celastrol significantly inhibited poly(I:C)-induced expression of adhesion molecules, such as ICAM-1/VCAM-1, and chemokines, such as CCL2, CXCL8, and CXCL10, in CRT-MG human astroglioma cells. In addition, celastrol significantly suppressed poly(I:C)-induced activation of JNK MAPK and STAT1 signaling pathways. Furthermore, celastrol significantly suppressed poly(I:C)-induced activation of the NF-κB signaling pathway. These results suggest that celastrol may exert its regulatory activity by inhibiting poly(I:C)-induced expression of pro-inflammatory mediators by suppressing activation of JNK MAPK-STAT1/NF-κB in astrocytes. [BMB Reports 2017; 50(1): 25-30].
Collapse
Affiliation(s)
- Soo Yeon An
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Gi Soo Youn
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Hyejin Kim
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Jinseu Park
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
9
|
Eriksen W. The spread of EBV to ectopic lymphoid aggregates may be the final common pathway in the pathogenesis of ME/CFS. Med Hypotheses 2017; 102:8-15. [PMID: 28478837 DOI: 10.1016/j.mehy.2017.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/02/2017] [Accepted: 02/26/2017] [Indexed: 12/22/2022]
Abstract
According to the hypothesis presented here, myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) develops over 3 steps: Step 1 is characterized by the aggregation of lymphoid cells in dorsal root ganglia or other nervous structures. The cause of this formation of ectopic lymphoid aggregates may be an acute infection, asymptomatic reactivations of a common neurotropic virus, exposure to a neurotoxin, or physical injury to peripheral nerves. In step 2, Epstein-Barr virus (EBV)-infected lymphocytes or monocytes bring EBV from the circulation to one or several of these lymphoid aggregates, whereupon cell-to-cell transmission of EBV and proliferation of latently EBV-infected lymphocytes lead to the presence of many EBV-infected cells in the lymphoid aggregates. The EBV-infected cells in the aggregates ignite an inflammation in the surrounding nervous tissue. This local inflammation elicits, in turn, a wave of glial cell activation that spreads from the EBV-infected area to parts of the nervous system that are not EBV-infected, disturbing the neuron-glial interaction in both the peripheral - and central nervous system. In step 3, immune cell exhaustion contributes to a consolidation of the pathological processes. There might be a cure: Infusions of autologous EBV-specific T-lymphocytes can perhaps remove the EBV-infected cells from the nervous system.
Collapse
Affiliation(s)
- Willy Eriksen
- Domain for Mental and Physical Health, Norwegian Institute of Public Health, Box 4404 Nydalen, 0403 Oslo, Norway.
| |
Collapse
|
10
|
Lisovska N, Daribayev Z, Lisovskyy Y, Kussainova K, Austin L, Bulekbayeva S. Pathogenesis of cerebral palsy through the prism of immune regulation of nervous tissue homeostasis: literature review. Childs Nerv Syst 2016; 32:2111-2117. [PMID: 27638717 DOI: 10.1007/s00381-016-3245-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 09/02/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND The cerebral palsy is highly actual issue of pediatrics, causing significant neurological disability. Though the great progress in the neuroscience has been recently achieved, the pathogenesis of cerebral palsy is still poorly understood. METHODS In this work, we reviewed available experimental and clinical data concerning the role of immune cells in pathogenesis of cerebral palsy. Maintaining of homeostasis in nervous tissue and its transformation in case of periventricular leukomalacia were analyzed. RESULTS The reviewed data demonstrate involvement of immune regulatory cells in the formation of nervous tissue imbalance and chronicity of inborn brain damage. The supported opinion, that periventricular leukomalacia is not a static phenomenon, but developing process, encourages our optimism about the possibility of its correction. CONCLUSIONS The further studies of changes of the nervous and immune systems in cerebral palsy are needed to create fundamentally new directions of the specific therapy and individual schemes of rehabilitation.
Collapse
Affiliation(s)
- Natalya Lisovska
- Republican Children's Rehabilitation Center, Turan str., 36, Astana, Kazakhstan, 010000.
| | - Zholtay Daribayev
- Republican Children's Rehabilitation Center, Turan str., 36, Astana, Kazakhstan, 010000
| | - Yevgeny Lisovskyy
- Republican Children's Rehabilitation Center, Turan str., 36, Astana, Kazakhstan, 010000
| | - Kenzhe Kussainova
- Republican Children's Rehabilitation Center, Turan str., 36, Astana, Kazakhstan, 010000
| | - Lana Austin
- Department of Pediatrics, Parirenyatwa Group of hospitals, Harare, Zimbabwe
| | - Sholpan Bulekbayeva
- Republican Children's Rehabilitation Center, Turan str., 36, Astana, Kazakhstan, 010000
| |
Collapse
|
11
|
Sun L, Wang X, Zhou Y, Zhou RH, Ho WZ, Li JL. Exosomes contribute to the transmission of anti-HIV activity from TLR3-activated brain microvascular endothelial cells to macrophages. Antiviral Res 2016; 134:167-171. [PMID: 27496004 DOI: 10.1016/j.antiviral.2016.07.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 07/01/2016] [Indexed: 12/17/2022]
Abstract
Human brain microvascular endothelial cells (HBMECs), the major cell type in the blood-brain barrier (BBB), play a key role in maintaining brain homeostasis. However, their role in the BBB innate immunity against HIV invasion of the central nervous system (CNS) remains to be determined. Our early work showed that TLR3 signaling of HBMECs could produce the antiviral factors that inhibit HIV replication in macrophages. The present study examined whether exosomes from TLR3-activated HBMECs mediate the intercellular transfer of antiviral factors to macrophages. Primary human macrophages could take up exosomes from TLR3-activated HBMECs. HBMECs-derived exosomes contained multiple antiviral factors, including several key IFN-stimulated genes (ISGs; ISG15, ISG56, and Mx2) at mRNA and protein levels. The depletion of exosomes from TLR3-activated HBMECs culture supernatant diminished HBMECs-mediated anti-HIV activity in macrophages. In conclusion, we demonstrate that exosomes shed by HBMECs are able to transport the antiviral molecules to macrophages. This finding suggests the possibility that HIV nonpermissive BBB cells (HBMECs) can help to restore the antiviral state in HIV-infected macrophages, which may be a defense mechanism against HIV neuroinvasion.
Collapse
Affiliation(s)
- Li Sun
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xu Wang
- State Key Laboratory of Virology, Wuhan University, Wuhan, 430071, China; Department of Pathology and Laboratory Medicine, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Yu Zhou
- Department of Pathology and Laboratory Medicine, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Run-Hong Zhou
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Wen-Zhe Ho
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China; State Key Laboratory of Virology, Wuhan University, Wuhan, 430071, China; Department of Pathology and Laboratory Medicine, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA.
| | - Jie-Liang Li
- Department of Pathology and Laboratory Medicine, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA.
| |
Collapse
|