1
|
Nazeen S, Wang X, Morrow A, Strom R, Ethier E, Ritter D, Henderson A, Afroz J, Stitziel NO, Gupta RM, Luk K, Studer L, Khurana V, Sunyaev SR. NERINE reveals rare variant associations in gene networks across multiple phenotypes and implicates an SNCA-PRL-LRRK2 subnetwork in Parkinson's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631688. [PMID: 39829934 PMCID: PMC11741352 DOI: 10.1101/2025.01.07.631688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Gene networks encapsulate biological knowledge, often linked to polygenic diseases. While model system experiments generate many plausible gene networks, validating their role in human phenotypes requires evidence from human genetics. Rare variants provide the most straightforward path for such validation. While single-gene analyses often lack power due to rare variant sparsity, expanding the unit of association to networks offers a powerful alternative, provided it integrates network connections. Here, we introduce NERINE, a hierarchical model-based association test that integrates gene interactions that integrates gene interactions while remaining robust to network inaccuracies. Applied to biobanks, NERINE uncovers compelling network associations for breast cancer, cardiovascular diseases, and type II diabetes, undetected by single-gene tests. For Parkinson's disease (PD), NERINE newly substantiates several GWAS candidate loci with rare variant signal and synergizes human genetics with experimental screens targeting cardinal PD pathologies: dopaminergic neuron survival and alpha-synuclein pathobiology. CRISPRi-screening in human neurons and NERINE converge on PRL , revealing an intraneuronal α-synuclein/prolactin stress response that may impact resilience to PD pathologies.
Collapse
|
2
|
Channer B, Matt SM, Nickoloff-Bybel EA, Pappa V, Agarwal Y, Wickman J, Gaskill PJ. Dopamine, Immunity, and Disease. Pharmacol Rev 2023; 75:62-158. [PMID: 36757901 PMCID: PMC9832385 DOI: 10.1124/pharmrev.122.000618] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/14/2022] Open
Abstract
The neurotransmitter dopamine is a key factor in central nervous system (CNS) function, regulating many processes including reward, movement, and cognition. Dopamine also regulates critical functions in peripheral organs, such as blood pressure, renal activity, and intestinal motility. Beyond these functions, a growing body of evidence indicates that dopamine is an important immunoregulatory factor. Most types of immune cells express dopamine receptors and other dopaminergic proteins, and many immune cells take up, produce, store, and/or release dopamine, suggesting that dopaminergic immunomodulation is important for immune function. Targeting these pathways could be a promising avenue for the treatment of inflammation and disease, but despite increasing research in this area, data on the specific effects of dopamine on many immune cells and disease processes remain inconsistent and poorly understood. Therefore, this review integrates the current knowledge of the role of dopamine in immune cell function and inflammatory signaling across systems. We also discuss the current understanding of dopaminergic regulation of immune signaling in the CNS and peripheral tissues, highlighting the role of dopaminergic immunomodulation in diseases such as Parkinson's disease, several neuropsychiatric conditions, neurologic human immunodeficiency virus, inflammatory bowel disease, rheumatoid arthritis, and others. Careful consideration is given to the influence of experimental design on results, and we note a number of areas in need of further research. Overall, this review integrates our knowledge of dopaminergic immunology at the cellular, tissue, and disease level and prompts the development of therapeutics and strategies targeted toward ameliorating disease through dopaminergic regulation of immunity. SIGNIFICANCE STATEMENT: Canonically, dopamine is recognized as a neurotransmitter involved in the regulation of movement, cognition, and reward. However, dopamine also acts as an immune modulator in the central nervous system and periphery. This review comprehensively assesses the current knowledge of dopaminergic immunomodulation and the role of dopamine in disease pathogenesis at the cellular and tissue level. This will provide broad access to this information across fields, identify areas in need of further investigation, and drive the development of dopaminergic therapeutic strategies.
Collapse
Affiliation(s)
- Breana Channer
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Stephanie M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Emily A Nickoloff-Bybel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Vasiliki Pappa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Yash Agarwal
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Jason Wickman
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Peter J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| |
Collapse
|
3
|
Khezri MR, Ghasemnejad-Berenji M. Icariin: A Potential Neuroprotective Agent in Alzheimer's Disease and Parkinson's Disease. Neurochem Res 2022; 47:2954-2962. [PMID: 35802286 DOI: 10.1007/s11064-022-03667-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 06/16/2022] [Accepted: 06/18/2022] [Indexed: 11/28/2022]
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the most common neurodegenerative diseases worldwide. They are characterized by the loss of neurons and synapses in special parts of the central nervous system (CNS). There is no definitive treatment for AD and PD, but extensive studies are underway to identify the effective drugs which can slow the progression of these diseases by affecting the factors involved in their pathophysiology (i.e., aggregated proteins, neuroinflammation, and oxidative stress). Icariin, a natural compound isolated from Epimedii herba, is known because of its anti-inflammatory and anti-oxidant properties. In this regard, there are numerous studies indicating its potential as a natural compound against the progression of CNS disorders, such as neurodegenerative diseases. Therefore, this review aims to re-examine findings on the pharmacologic effects of icariin on factors involved in the pathophysiology of AD and PD.
Collapse
Affiliation(s)
| | - Morteza Ghasemnejad-Berenji
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran. .,Research Center for Experimental and Applied Pharmaceutical Sciences, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
4
|
Dolatshahi M, Ranjbar Hameghavandi MH, Sabahi M, Rostamkhani S. Nuclear factor-kappa B (NF-κB) in pathophysiology of Parkinson disease: Diverse patterns and mechanisms contributing to neurodegeneration. Eur J Neurosci 2021; 54:4101-4123. [PMID: 33884689 DOI: 10.1111/ejn.15242] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/25/2021] [Accepted: 04/07/2021] [Indexed: 01/02/2023]
Abstract
Parkinson's disease (PD), the most common movement disorder, comprises several pathophysiologic mechanisms including misfolded alpha-synuclein aggregation, inflammation, mitochondrial dysfunction, and synaptic loss. Nuclear Factor-Kappa B (NF-κB), as a key regulator of a myriad of cellular reactions, is shown to be involved in such mechanisms associated with PD, and the changes in NF-κB expression is implicated in PD. Alpha-synuclein accumulation, the characteristic feature of PD pathology, is known to trigger NF-κB activation in neurons, thereby propagating apoptosis through several mechanisms. Furthermore, misfolded alpha-synuclein released from degenerated neurons, activates several signaling pathways in glial cells which culminate in activation of NF-κB and production of pro-inflammatory cytokines, thereby aggravating neurodegenerative processes. On the other hand, NF-κB activation, acting as a double-edged sword, can be necessary for survival of neurons. For instance, NF-κB activation is necessary for competent mitochondrial function and deficiency in c-Rel, one of the NF-κB proteins, is known to propagate DA neuron loss via several mechanisms. Despite the dual role of NF-κB in PD, several agents by selectively modifying the mechanisms and pathways associated with NF-κB, can be effective in attenuating DA neuron loss and PD, as reviewed in this paper.
Collapse
Affiliation(s)
- Mahsa Dolatshahi
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | | | - Mohammadmahdi Sabahi
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Neurosurgery Research Group (NRG), Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sabra Rostamkhani
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
RNF11 at the Crossroads of Protein Ubiquitination. Biomolecules 2020; 10:biom10111538. [PMID: 33187263 PMCID: PMC7697665 DOI: 10.3390/biom10111538] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/06/2020] [Accepted: 11/08/2020] [Indexed: 12/18/2022] Open
Abstract
RNF11 (Ring Finger Protein 11) is a 154 amino-acid long protein that contains a RING-H2 domain, whose sequence has remained substantially unchanged throughout vertebrate evolution. RNF11 has drawn attention as a modulator of protein degradation by HECT E3 ligases. Indeed, the large number of substrates that are regulated by HECT ligases, such as ITCH, SMURF1/2, WWP1/2, and NEDD4, and their role in turning off the signaling by ubiquitin-mediated degradation, candidates RNF11 as the master regulator of a plethora of signaling pathways. Starting from the analysis of the primary sequence motifs and from the list of RNF11 protein partners, we summarize the evidence implicating RNF11 as an important player in modulating ubiquitin-regulated processes that are involved in transforming growth factor beta (TGF-β), nuclear factor-κB (NF-κB), and Epidermal Growth Factor (EGF) signaling pathways. This connection appears to be particularly significant, since RNF11 is overexpressed in several tumors, even though its role as tumor growth inhibitor or promoter is still controversial. The review highlights the different facets and peculiarities of this unconventional small RING-E3 ligase and its implication in tumorigenesis, invasion, neuroinflammation, and cancer metastasis.
Collapse
|
6
|
Privman Champaloux E, Donelson N, Pyakurel P, Wolin D, Ostendorf L, Denno M, Borman R, Burke C, Short-Miller JC, Yoder MR, Copeland JM, Sanyal S, Venton BJ. Ring Finger Protein 11 (RNF11) Modulates Dopamine Release in Drosophila. Neuroscience 2020; 452:37-48. [PMID: 33176188 DOI: 10.1016/j.neuroscience.2020.10.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/15/2020] [Accepted: 10/18/2020] [Indexed: 02/06/2023]
Abstract
Recent work indicates a role for RING finger protein 11 (RNF11) in Parkinson disease (PD) pathology, which involves the loss of dopaminergic neurons. However, the role of RNF11 in regulating dopamine neurotransmission has not been studied. In this work, we tested the effect of RNF11 RNAi knockdown or overexpression on stimulated dopamine release in the larval Drosophila central nervous system. Dopamine release was stimulated using optogenetics and monitored in real-time using fast-scan cyclic voltammetry at an electrode implanted in an isolated ventral nerve cord. RNF11 knockdown doubled dopamine release, but there was no decrease in dopamine from RNF11 overexpression. RNF11 knockdown did not significantly increase stimulated serotonin or octopamine release, indicating the effect is dopamine specific. Dopamine clearance was also changed, as RNF11 RNAi flies had a higher Vmax and RNF11 overexpressing flies had a lower Vmax than control flies. RNF11 RNAi flies had increased mRNA levels of dopamine transporter (DAT) in RNF11, confirming changes in DAT. In RNF11 RNAi flies, release was maintained better for stimulations repeated at short intervals, indicating increases in the recycled releasable pool of dopamine. Nisoxetine, a DAT inhibitor, and flupenthixol, a D2 antagonist, did not affect RNF11 RNAi or overexpressing flies differently than control. Thus, RNF11 knockdown causes early changes in dopamine neurotransmission, and this is the first work to demonstrate that RNF11 affects both dopamine release and uptake. RNF11 expression decreases in human dopaminergic neurons during PD, and that decrease may be protective by increasing dopamine neurotransmission in the surviving dopaminergic neurons.
Collapse
Affiliation(s)
- Eve Privman Champaloux
- Neuroscience Graduate Program and Medical Scientist Training Program, University of Virginia, Charlottesville, VA, United States
| | | | - Poojan Pyakurel
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Danielle Wolin
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Leah Ostendorf
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Madelaine Denno
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Ryan Borman
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | | | - Jonah C Short-Miller
- Department of Biology, Eastern Mennonite University, Harrisonburg, VA, United States
| | - Maria R Yoder
- Department of Biology, Eastern Mennonite University, Harrisonburg, VA, United States
| | - Jeffrey M Copeland
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States; Department of Biology, Eastern Mennonite University, Harrisonburg, VA, United States
| | | | - B Jill Venton
- Neuroscience Graduate Program and Medical Scientist Training Program, University of Virginia, Charlottesville, VA, United States; Department of Chemistry, University of Virginia, Charlottesville, VA, United States.
| |
Collapse
|
7
|
Jing N, Li X. Dihydromyricetin Attenuates Inflammation through TLR4/NF-kappaB Pathway. Open Med (Wars) 2019; 14:719-725. [PMID: 31572805 PMCID: PMC6749725 DOI: 10.1515/med-2019-0083] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 07/29/2019] [Indexed: 12/14/2022] Open
Abstract
Microglia plays a complex role in neuroinflammation, which has been implicated in neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. This study aims to explore the effect and mechanism of Dihydromyricetin (DHM) on lipopolysaccharide (LPS)-induced inflammation in microglial BV-2 cells. Cell viability was measured by 3-[4,5-dimethylthiazol-2-yl]2,5-diphenyltetrazolium bromide (MTT) assay. The pro-inflammatory mediators and cytokines including interleukin (IL)-6, IL-1β, and tumor necrosis factor-α (TNF-α); inducible nitric oxide synthase (iNOS); and cyclooxygenase 2 (COX-2) were measured by enzyme-linked immunosorbent assay (ELISA) and/or quantitative real-time PCR (qRT-PCR). The expression of p-p65, p-IκBα, toll-like receptor 4 (TLR4), and myeloid differentiation primary response 88 (MyD88) were analyzed by western blot. The present study showed that DHM treatment alleviated LPS-induced viability reduction, suppressed the mRNA levels of IL-6, IL-1β and TNF-α, inhibited the mRNA and protein expression of iNOS and COX-2, and attenuated the activation of NF-кB and TLR4 signaling in a concentration-dependent manner. In conclusion, DHM exerts an anti-inflammatory effect on LPS-induced BV-2 microglial cells, possibly through TRL4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Nianshui Jing
- Department of Pharmacy, Jinan No.2 People's Hospital, Jingyi Road 148, Jinan 250001, Shandong, China
| | - Xinnan Li
- Department of Pharmacy, Jinan No.2 People's Hospital, Jingyi Road 148, Jinan 250001, Shandong, China
| |
Collapse
|
8
|
Zhang J, Liu S, Zhang D, Ma Z, Sun L. Homeobox D10, a tumor suppressor, inhibits the proliferation and migration of esophageal squamous cell carcinoma. J Cell Biochem 2019; 120:13717-13725. [PMID: 30938888 DOI: 10.1002/jcb.28644] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/31/2019] [Accepted: 02/04/2019] [Indexed: 12/18/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most common types of esophageal cancer, which is the sixth leading cause of cancer death globally. Homeobox D10 (HOXD10) is a member of the homeobox (HOX) gene family and has been reported to act as a tumor suppressor. However, the potential role of HOXD10 in ESCC has not been reported. Thus, the aim of this study was to examine the expression and function of HOXD10 in ESCC. The expressions of HOXD10 in human ESCC tissues and cell lines were detected by quantitative reverse transcription polymerase chain reaction and Western blot. The HOXD10 overexpressing cell lines were established, then CCK-8 and Transwell assays were performed to examine cell proliferation, migration, and invasion, respectively. The expression of EMT-related proteins and signaling pathway-related proteins were detected by Western blot. Our results showed that HOXD10 is lowly expressed in ESCC tissues as well as in ESCC cell lines. Ectopic overexpression of HOXD10 inhibited cell proliferation, migration, and invasion of ESCC cells (P < 0.05). HOXD10 overexpression repressed the epithelial-mesenchymal transition (EMT) process in ESCC cells. Besides, HOXD10 overexpression suppressed the activation of PI3K/AKT/mTOR signaling pathway. PI3K/Akt agonist, insulin-like growth factor-1, reversed the inhibitory effects of HOXD10 on cell proliferation and migration in ESCC cells. Additional in vivo study proved that ectopic expression of HOXD10 caused an obvious inhibitory effect on the tumor growth. These findings indicated that overexpression of HOXD10 suppressed the proliferation, migration, and invasion via regulating the PI3K/AKT/mTOR signaling pathway in ESCC cells. Thus, targeting HOXD10 may be considered as a therapeutic strategy for ESCC treatment.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Shiyuan Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Danjie Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhenchuan Ma
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Liangzhang Sun
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
9
|
Gorelenkova Miller O, Mieyal JJ. Critical Roles of Glutaredoxin in Brain Cells-Implications for Parkinson's Disease. Antioxid Redox Signal 2019; 30:1352-1368. [PMID: 29183158 PMCID: PMC6391617 DOI: 10.1089/ars.2017.7411] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Glutaredoxin (Grx)1, an evolutionarily conserved and ubiquitous enzyme, regulates redox signal transduction and protein redox homeostasis by catalyzing reversible S-glutathionylation. Grx1 plays different roles in different cell types. In Parkinson's disease (PD), Grx1 regulates apoptosis signaling in dopaminergic neurons, so that loss of Grx1 leads to increased cell death; in microglial cells, Grx1 regulates proinflammatory signaling, so that upregulation of Grx1 promotes cytokine production. Here we examine the regulatory roles of Grx1 in PD with a view toward therapeutic innovation. Recent Advances: In postmortem midbrain PD samples, Grx1 was decreased relative to controls, specifically within dopaminergic neurons. In Caenorhabditis elegans models of PD, loss of the Grx1 homologue led to exacerbation of the neurodegenerative phenotype. This effect was partially relieved by overexpression of neuroprotective DJ-1, consistent with regulation of DJ-1 content by Grx1. Increased GLRX copy number in PD patients was associated with earlier PD onset; and Grx1 levels correlated with levels of proinflammatory tumor necrosis factor-α in mouse and human brain samples. In vitro studies showed Grx1 to be upregulated on proinflammatory activation of microglia. Direct overexpression of Grx1 increased microglial activation; silencing Grx1 diminished activation. Grx1 upregulation in microglia corresponded to increased neuronal cell death in coculture. Overall, these studies identify competing roles of Grx1 in PD etiology. CRITICAL ISSUES The dilemma regarding Grx1 as a PD therapeutic target is whether to stimulate its upregulation for neuroprotection or inhibit its proinflammatory activity. FUTURE DIRECTIONS Further investigation is needed to understand the preponderant role of Grx1 regarding dopaminergic neuronal survival.
Collapse
Affiliation(s)
- Olga Gorelenkova Miller
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - John J Mieyal
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
10
|
Cui B, Guo X, You Y, Fu R. Farrerol attenuates MPP + -induced inflammatory response by TLR4 signaling in a microglia cell line. Phytother Res 2019; 33:1134-1141. [PMID: 30734970 DOI: 10.1002/ptr.6307] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 12/17/2018] [Accepted: 01/11/2019] [Indexed: 12/13/2022]
Abstract
Farrerol was found to possess neuroprotective effect; however, the mechanism remains unknown. The aim of the present study was to explore the effect of farrerol on MPP+ -induced inflammation in mouse microglial BV-2 cells and to elaborate the underlying mechanism. MTT assay was performed to measure the cell viability. The pro-inflammatory mediators and cytokines including interleukin (IL)-6, IL-1β, and tumor necrosis factor-α (TNF-α); inducible nitric oxide synthase; and cyclooxygenase 2 were measured. The expression of p-p65, p-IκBα, toll-like receptor 4 (TLR4), and myeloid differentiation primary response 88 were analyzed by western blot. We found that farrerol treatment improved cell viability in MPP+ -induced BV-2 cells. MPP+ -induced upregulation of IL-6, IL-1β, and TNF-α was inhibited by farrerol treatment. Farrerol treatment also attenuated MPP+ -induced expression of inducible nitric oxide synthase and cyclooxygenase 2 as well as the activation of NF-κB in BV-2 cells. MPP+ -induced TLR4 signaling was markedly diminished by farrerol treatment. Knockdown of TLR4 attenuated MPP+ -induced inflammatory response in BV-2 cells. In conclusion, farrerol treatment attenuated MPP+ -induced inflammatory response by inhibiting the TLR4 signaling pathway in BV-2 cells. The results indicated that farrerol could be used as a therapeutic agent for preventing or alleviating the neuroinflammation-related diseases, such as Parkinson's disease.
Collapse
Affiliation(s)
- Bei Cui
- Department of Neurology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Xiaqing Guo
- Department of Neurology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Yanhui You
- Department of Neurology, Tongxu County Hospital, Kaifeng, China
| | - Rongli Fu
- Department of Neurology, Huaihe Hospital of Henan University, Kaifeng, China
| |
Collapse
|
11
|
Gao Y, Zhong J, Jiang L. Raf kinase inhibitor protein protects microglial cells against 1-methyl-4-phenylpyridinium-induced neuroinflammation in vitro. Exp Cell Res 2018; 372:108-117. [PMID: 30244177 DOI: 10.1016/j.yexcr.2018.09.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 09/15/2018] [Accepted: 09/20/2018] [Indexed: 11/17/2022]
Abstract
The Raf kinase inhibitor protein (RKIP), belonging to a member of the phosphatidylethanolamine-binding protein (PEBP) family, is involved in regulating neural development. However, the role of RKIP in microglial cells stimulated with 1-methyl-4-phenylpyridinium (MPP+) has not been determined. Thus, in the present study, we investigated the role of RKIP and its underlying mechanism in Parkinson's disease (PD). Our results showed that the expression of RKIP was significantly reduced in BV-2 cells treated with MPP+. Overexpression of RKIP markedly rescued cell viability and inhibited cell apoptosis in BV-2 cells exposed to MPP+. In addition, overexpression of RKIP inhibited MPP+-induced the production of pro-inflammatory molecules in BV-2 cells. Similar results were observed in primary microglial cells isolated from neonatal mice. Exploration of the underlying mechanisms of its action indicated that overexpression of RKIP prevented the activation of NF-κB and MEK/ERK pathways in MPP+-stimulated BV-2 cells. Taken together, these findings indicated that RKIP suppresses apoptosis and inflammation in MPP+-treated microglial cells through the inactivation of NF-κB and MEK/ERK signaling pathways. Thus, RKIP may be a promising target molecular involving in the pathogenesis of PD.
Collapse
Affiliation(s)
- Yuanlin Gao
- The First ward of Neurology Department, Kaifeng Central Hospital, Kaifeng 475000, Henan Province, China
| | - Jie Zhong
- Department of Nursing, Kaifeng Central Hospital, Kaifeng 475000, Henan Province, China.
| | - Lei Jiang
- The First ward of Neurology Department, Kaifeng Central Hospital, Kaifeng 475000, Henan Province, China
| |
Collapse
|
12
|
Model Senescent Microglia Induce Disease Related Changes in α-Synuclein Expression and Activity. Biomolecules 2018; 8:biom8030067. [PMID: 30071596 PMCID: PMC6164966 DOI: 10.3390/biom8030067] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/24/2018] [Accepted: 07/26/2018] [Indexed: 12/13/2022] Open
Abstract
Aging is the most prominent risk factor for most neurodegenerative diseases. However, incorporating aging-related changes into models of neurodegeneration rarely occurs. One of the significant changes that occurs in the brain as we age is the shift in phenotype of the resident microglia population to one less able to respond to deleterious changes in the brain. These microglia are termed dystrophic microglia. In order to better model neurodegenerative diseases, we have developed a method to convert microglia into a senescent phenotype in vitro. Mouse microglia grown in high iron concentrations showed many characteristics of dystrophic microglia including, increased iron storage, increased expression of proteins, such as ferritin and the potassium channel, Kv1.3, increased reactive oxygen species production and cytokine release. We have applied this new model to the study of α-synuclein, a protein that is closely associated with a number of neurodegenerative diseases. We have shown that conditioned medium from our model dystrophic microglia increases α-synuclein transcription and expression via tumor necrosis factor alpha (TNFα) and mediated through nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). The conditioned medium also decreases the formation of α-synuclein tetramers, associated ferrireductase activity, and increases aggregates of α-synuclein. The results suggest that we have developed an interesting new model of aged microglia and that factors, including TNFα released from dystrophic microglia could have a significant influence on the pathogenesis of α-synuclein related diseases.
Collapse
|
13
|
Kempuraj D, Thangavel R, Selvakumar GP, Ahmed ME, Zaheer S, Raikwar SP, Zahoor H, Saeed D, Dubova I, Giler G, Herr S, Iyer SS, Zaheer A. Mast Cell Proteases Activate Astrocytes and Glia-Neurons and Release Interleukin-33 by Activating p38 and ERK1/2 MAPKs and NF-κB. Mol Neurobiol 2018; 56:1681-1693. [PMID: 29916143 DOI: 10.1007/s12035-018-1177-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/04/2018] [Indexed: 12/13/2022]
Abstract
Inflammatory mediators released from activated microglia, astrocytes, neurons, and mast cells mediate neuroinflammation. Parkinson's disease (PD) is characterized by inflammation-dependent dopaminergic neurodegeneration in substantia nigra. 1-Methyl-4-phenylpyridinium (MPP+), a metabolite of parkinsonian neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), induces inflammatory mediators' release from brain cells and mast cells. Brain cells' interaction with mast cells is implicated in neuroinflammation. However, the exact mechanisms involved are not yet clearly understood. Mouse fetal brain-derived cultured primary astrocytes and glia-neurons were incubated with mouse mast cell protease-6 (MMCP-6) and MMCP-7, and mouse bone marrow-derived mast cells (BMMCs) were incubated with MPP+ and brain protein glia maturation factor (GMF). Interleukin-33 (IL-33) released from these cells was quantitated by enzyme-linked immunosorbent assay. Both MMCP-6 and MMCP-7 induced IL-33 release from astrocytes and glia-neurons. MPP+ and GMF were used as a positive control-induced IL-33 and reactive oxygen species expression in mast cells. Mast cell proteases and MPP+ activate p38 and extracellular signal-regulated kinases 1/2 (ERK1/2), mitogen-activated protein kinases (MAPKs), and transcription factor nuclear factor-kappa B (NF-κB) in astrocytes, glia-neurons, or mast cells. Addition of BMMCs from wt mice and transduction with adeno-GMF show higher chemokine (C-C motif) ligand 2 (CCL2) release. MPP+ activated glial cells and reduced microtubule-associated protein 2 (MAP-2) expression indicating neurodegeneration. IL-33 expression increased in the midbrain and striatum of PD brains as compared with age- and sex-matched control subjects. Glial cells and neurons interact with mast cells and accelerate neuroinflammation and these interactions can be explored as a new therapeutic target to treat PD.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA.
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA.
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Gvindhasamy Pushpavathi Selvakumar
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Mohammad Ejaz Ahmed
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Smita Zaheer
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Haris Zahoor
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Daniyal Saeed
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Iuliia Dubova
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Gema Giler
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Shelby Herr
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Shankar S Iyer
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA
| | - Asgar Zaheer
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, MO, USA.
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Medical Science Building,1 Hospital Drive, Columbia, MO, 65211, USA.
| |
Collapse
|
14
|
Budhidarmo R, Zhu J, Middleton AJ, Day CL. The RING domain of RING Finger 11 (RNF11) protein binds Ubc13 and inhibits formation of polyubiquitin chains. FEBS Lett 2018. [PMID: 29537486 DOI: 10.1002/1873-3468.13029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The Really Interesting New Gene (RING) Finger protein 11 (RNF11) is a subunit of the A20 ubiquitin-editing complex that ensures the transient nature of inflammatory responses. Although the role of RNF11 as a negative regulator of NF-κB signalling is well-documented, the molecular mechanisms that underpin this function are poorly understood. Here, we show that RNF11 binds both Ubc13 and the Ubc13~ubiquitin conjugate tightly and with similar affinity, but has minimal E3 ligase activity. Remarkably, RNF11 appears to bind Ubc13 so tightly that it outcompetes the E1 and an active E3 ligase. As a consequence, RNF11 may regulate the activity of E3s that rely on Ubc13 for ubiquitin chain assembly by limiting the availability of Ubc13 and its conjugate.
Collapse
Affiliation(s)
- Rhesa Budhidarmo
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Jingyi Zhu
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Adam J Middleton
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Catherine L Day
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
15
|
Liu H, Yu C, Xu T, Zhang X, Dong M. Synergistic protective effect of paeoniflorin and β-ecdysterone against rotenone-induced neurotoxicity in PC12 cells. Apoptosis 2018; 21:1354-1365. [PMID: 27688248 DOI: 10.1007/s10495-016-1293-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
There are several factors, like oxidative stress and neurons loss, involving neurodegenerative diseases such as Parkinson's disease (PD). The combination of antioxidant and anti-apoptotic agent is becoming a promising approach to fight against PD. This study evaluates the hypothesis that paeoniflorin (PF) and β-ecdysterone (β-Ecd) synergize to protect PC12 cells against toxicity induced by PD-related neurotoxin rotenone. The combination of PF and β-Ecd, hereafter referred to as the PF/β-Ecd, at suboptimal concentrations increased the viability of rotenone-exposed PC12 cells in a synergistic manner. PF and β-Ecd cooperate to attenuate the rotenone-induced apoptosis by decrease in Bax expression, caspase-9 activity, and caspase-3 activity. PF or PF/β-Ecd, but not β-Ecd, inhibited rotenone-triggered protein kinase C-δkinase C-δ (PKCδ) upregulation and nuclear factor κB (NF-κB) activation. β-Ecd or PF/β-Ecd, but not PF, enhanced serine/threonine protein kinase (Akt) activation, promoted nuclear factor E2-related factor 2 (Nrf2) nuclear accumulation, suppressed reactive oxygen species (ROS) production. Neuroprotection of PF/β-Ecd could be completely blocked by PKCδ inhibitor rottlerin plus Akt specific inhibitor LY294002. Dual blockade of the PKCδ/NF-κB pathway by PF and activation of Akt/Nrf2 pathway by β-Ecd results in a synergistic neuroprotective effect against rotenone-induced neurotoxicity in vitro. These findings provide the rationale for determining the in vivo activity of combined therapy with PF and β-Ecd against PD.
Collapse
Affiliation(s)
- Han Liu
- The Institute of Medicine, Qiqihar Medical University, 333 BuKui Street, JianHua District, Qiqihar, 161006, China
| | - Chunlei Yu
- The Institute of Medicine, Qiqihar Medical University, 333 BuKui Street, JianHua District, Qiqihar, 161006, China
| | - Tianjiao Xu
- The Institute of Medicine, Qiqihar Medical University, 333 BuKui Street, JianHua District, Qiqihar, 161006, China
| | - Xiaojie Zhang
- The Institute of Medicine, Qiqihar Medical University, 333 BuKui Street, JianHua District, Qiqihar, 161006, China
| | - Miaoxian Dong
- The Institute of Medicine, Qiqihar Medical University, 333 BuKui Street, JianHua District, Qiqihar, 161006, China.
| |
Collapse
|
16
|
Tian K, Lan X, Zhu Q, Liu LB, Yin HD, Zhao XL, Li DY, Zhang Y, Wang Y. RNF11 negatively regulates antiviral signaling and incites inflammatory response in chicken embryo fibroblast. EUR J INFLAMM 2017. [DOI: 10.1177/1721727x17742547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RNF11 (RING finger protein 11) has been indicated to be related to numerous immunological diseases. As a neo-regulator in innate immunity, RNF11 was proved to inhibit antiviral signaling and interferon-β (IFN-β) production in human cell line. Chicken antiviral signaling pathway is different with other species. However, there is little study about the role of RNF11 in chicken antiviral signaling pathway and inflammatory response. This study was designed to identify the function of RNF11 in chicken antiviral signaling pathway and inflammatory response. We overexpressed and interfered RNF11 in chicken embryo fibroblasts (CEFs) and then used quantitative reverse transcription polymerase chain reaction (qRT-PCR) to investigate the poly (I:C)-induced production of IFN-β and inflammatory response factors IL2 and IL10. The results indicated that RNF11 extremely significantly decreased the production of IFN-β and highly significantly increased the messenger RNA (mRNA) levels of IL2 and IL10, but depletion of RNF11 obtained inverse results. Thus, we concluded that RNF11 not only can inhibit antiviral signaling but also plays an important role in inflammatory response in chicken.
Collapse
Affiliation(s)
- Kai Tian
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Xi Lan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Qing Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Ling-Bin Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Hua-Dong Yin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Xiao-Ling Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Di-Yan Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Yao Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu Campus, Chengdu, China
| |
Collapse
|
17
|
A20 in Multiple Sclerosis and Parkinson’s Disease: Clue to a Common Dysregulation of Anti-Inflammatory Pathways? Neurotox Res 2017; 32:1-7. [DOI: 10.1007/s12640-017-9724-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 03/02/2017] [Accepted: 03/08/2017] [Indexed: 01/29/2023]
|
18
|
Souza RB, Frota AF, Sousa RS, Cezario NA, Santos TB, Souza LMF, Coura CO, Monteiro VS, Cristino Filho G, Vasconcelos SMM, da Cunha RMS, Aguiar LMV, Benevides NMB. Neuroprotective Effects of Sulphated Agaran from Marine Alga Gracilaria cornea in Rat 6-Hydroxydopamine Parkinson's Disease Model: Behavioural, Neurochemical and Transcriptional Alterations. Basic Clin Pharmacol Toxicol 2016; 120:159-170. [PMID: 27612165 DOI: 10.1111/bcpt.12669] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 08/31/2016] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a multifactorial disease associated with the degeneration of dopaminergic neurons and behavioural alterations. Natural bioactive compounds may provide new therapeutic alternatives for neurodegenerative disorders, such as PD. The sulphated polysaccharides isolated from marine algae are heterogenic molecules that show different biological activities. The red marine alga Gracilaria cornea has a sulphated polysaccharide (SA-Gc) with structure and anti-inflammatory and antinociceptive activities reported in the literature. Therefore, this study aimed to evaluate the neuroprotective effects of SA-Gc in rat model PD induced by 6-hydroxydopamine (6-OHDA). Firstly, we established the PD model in rats, induced by an intrastriatal injection (int.) of 6-OHDA, followed by a single administration of SA-Gc (15, 30 or 60 μg; int.). On the 14th day, behavioural tests were performed. After killing, brain areas were dissected and used for neurochemical and/or transcriptional analyses. The results showed that SA-Gc (60 μg, int.) promoted neuroprotective effects in vivo through reducing the oxidative/nitroactive stress and through alterations in the monoamine contents induced by 6-OHDA. Furthermore, SA-Gc modulated the transcription of neuroprotective and inflammatory genes, as well as returning behavioural activities and weight gain to normal conditions. Thus, this study reports the neuroprotective effects of SA-Gc against 6-OHDA in rats.
Collapse
Affiliation(s)
- Ricardo Basto Souza
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | | | | | | | | | | | - Chistiane Oliveira Coura
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Valdécio Silvano Monteiro
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | | | | | | | | | | |
Collapse
|
19
|
MicroRNA-19b-3p Modulates Japanese Encephalitis Virus-Mediated Inflammation via Targeting RNF11. J Virol 2016; 90:4780-4795. [PMID: 26937036 PMCID: PMC4836334 DOI: 10.1128/jvi.02586-15] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 02/18/2016] [Indexed: 01/16/2023] Open
Abstract
UNLABELLED Japanese encephalitis virus (JEV) can invade the central nervous system and consequently induce neuroinflammation, which is characterized by profound neuronal cell damage accompanied by astrogliosis and microgliosis. Albeit microRNAs (miRNAs) have emerged as major regulatory noncoding RNAs with profound effects on inflammatory response, it is unknown how astrocytic miRNAs regulate JEV-induced inflammation. Here, we found the involvement of miR-19b-3p in regulating the JEV-induced inflammatory responsein vitroandin vivo The data demonstrated that miR-19b-3p is upregulated in cultured cells and mouse brain tissues during JEV infection. Overexpression of miR-19b-3p led to increased production of inflammatory cytokines, including tumor necrosis factor alpha, interleukin-6, interleukin-1β, and chemokine (C-C motif) ligand 5, after JEV infection, whereas knockdown of miR-19b-3p had completely opposite effects. Mechanistically, miR-19b-3p modulated the JEV-induced inflammatory response via targeting ring finger protein 11, a negative regulator of nuclear factor kappa B signaling. We also found that inhibition of ring finger protein 11 by miR-19b-3p resulted in accumulation of nuclear factor kappa B in the nucleus, which in turn led to higher production of inflammatory cytokines.In vivosilencing of miR-19b-3p by a specific antagomir reinvigorates the expression level of RNF11, which in turn reduces the production of inflammatory cytokines, abrogates gliosis and neuronal cell death, and eventually improves the survival rate in the mouse model. Collectively, our results demonstrate that miR-19b-3p positively regulates the JEV-induced inflammatory response. Thus, miR-19b-3p targeting may constitute a thought-provoking approach to rein in JEV-induced inflammation. IMPORTANCE Japanese encephalitis virus (JEV) is one of the major causes of acute encephalitis in humans worldwide. The pathological features of JEV-induced encephalitis are inflammatory reactions and neurological diseases resulting from glia activation. MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene expression posttranscriptionally. Accumulating data indicate that miRNAs regulate a variety of cellular processes, including the host inflammatory response under pathological conditions. Recently, a few studies demonstrated the role of miRNAs in a JEV-induced inflammatory response in microglia; however, their role in an astrocyte-derived inflammatory response is largely unknown. The present study reveals that miR-19b-3p targets ring finger protein 11 in glia and promotes inflammatory cytokine production by enhancing nuclear factor kappa B activity in these cells. Moreover, administration of an miR-19b-3p-specific antagomir in JEV-infected mice reduces neuroinflammation and lethality. These findings suggest a new insight into the molecular mechanism of the JEV-induced inflammatory response and provide a possible therapeutic entry point for treating viral encephalitis.
Collapse
|
20
|
Abbasi A, Forsberg K, Bischof F. The role of the ubiquitin-editing enzyme A20 in diseases of the central nervous system and other pathological processes. Front Mol Neurosci 2015; 8:21. [PMID: 26124703 PMCID: PMC4466442 DOI: 10.3389/fnmol.2015.00021] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/24/2015] [Indexed: 11/28/2022] Open
Abstract
In recent years, the ubiquitin-editing enzyme A20 has been shown to control a large set of molecular pathways involved in the regulation of protective as well as self-directed immune responses. Here, we assess the current and putative roles of A20 in inflammatory, vascular and degenerative diseases of the central nervous system and explore future directions of research.
Collapse
Affiliation(s)
- Asghar Abbasi
- Department of Neuroimmunology, Hertie Institute for Clinical Brain Research and Center of Neurology, University Hospital Tübingen Tübingen, Germany
| | - Kirsi Forsberg
- Department of Neuroimmunology, Hertie Institute for Clinical Brain Research and Center of Neurology, University Hospital Tübingen Tübingen, Germany
| | - Felix Bischof
- Department of Neuroimmunology, Hertie Institute for Clinical Brain Research and Center of Neurology, University Hospital Tübingen Tübingen, Germany
| |
Collapse
|
21
|
Lanzillotta A, Porrini V, Bellucci A, Benarese M, Branca C, Parrella E, Spano PF, Pizzi M. NF-κB in Innate Neuroprotection and Age-Related Neurodegenerative Diseases. Front Neurol 2015; 6:98. [PMID: 26042083 PMCID: PMC4438602 DOI: 10.3389/fneur.2015.00098] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 04/21/2015] [Indexed: 12/20/2022] Open
Abstract
NF-κB factors are cardinal transcriptional regulators of inflammation and apoptosis, involved in the brain programing of systemic aging and in brain damage. The composition of NF-κB active dimers and epigenetic mechanisms modulating histone acetylation, finely condition neuronal resilience to brain insults. In stroke models, the activation of NF-κB/c-Rel promotes neuroprotective effects by transcription of specific anti-apoptotic genes. Conversely, aberrant activation of NF-κB/RelA showing reduced level of total acetylation, but site-specific acetylation on lysine 310, triggers the expression of pro-apoptotic genes. Constitutive knockout of c-Rel shatters the resilience of substantia nigra (SN) dopaminergic (DA) neurons to aging and induces a parkinsonian like pathology in mice. c-rel(-/-) mice show increased level of aberrantly acetylated RelA in the basal ganglia, neuroinflammation, accumulation of alpha-synuclein, and iron. Moreover, they develop motor deficits responsive to l-DOPA treatment and associated with loss of DA neurons in the SN. Here, we discuss the effect of unbalanced activation of RelA and c-Rel during aging and propose novel challenges for the development of therapeutic strategies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Annamaria Lanzillotta
- Department of Molecular and Translational Medicine, National Institute of Neuroscience, University of Brescia, Brescia, Italy
| | - Vanessa Porrini
- Department of Molecular and Translational Medicine, National Institute of Neuroscience, University of Brescia, Brescia, Italy
- IRCCS, San Camillo Hospital, Venice, Italy
| | - Arianna Bellucci
- Department of Molecular and Translational Medicine, National Institute of Neuroscience, University of Brescia, Brescia, Italy
| | - Marina Benarese
- Department of Molecular and Translational Medicine, National Institute of Neuroscience, University of Brescia, Brescia, Italy
| | - Caterina Branca
- Department of Molecular and Translational Medicine, National Institute of Neuroscience, University of Brescia, Brescia, Italy
| | - Edoardo Parrella
- Department of Molecular and Translational Medicine, National Institute of Neuroscience, University of Brescia, Brescia, Italy
| | - Pier Franco Spano
- Department of Molecular and Translational Medicine, National Institute of Neuroscience, University of Brescia, Brescia, Italy
- IRCCS, San Camillo Hospital, Venice, Italy
| | - Marina Pizzi
- Department of Molecular and Translational Medicine, National Institute of Neuroscience, University of Brescia, Brescia, Italy
- IRCCS, San Camillo Hospital, Venice, Italy
| |
Collapse
|
22
|
Yan J, Fu Q, Cheng L, Zhai M, Wu W, Huang L, Du G. Inflammatory response in Parkinson's disease (Review). Mol Med Rep 2014; 10:2223-33. [PMID: 25215472 DOI: 10.3892/mmr.2014.2563] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 07/01/2014] [Indexed: 11/05/2022] Open
Abstract
Parkinson's disease (PD) is one of the most common age‑related neurodegenerative diseases, which results from a number of environmental and inherited factors. PD is characterized by the slow progressive degeneration of dopaminergic (DA) neurons in the substantia nigra. The nigrostriatal DA neurons are particularly vulnerable to inflammatory attack. Neuroinflammation is an important contributor to the pathogenesis of age‑related neurodegenerative disorders, such as PD, and as such anti‑inflammatory agents are becoming a novel therapeutic focus. This review will discuss the current knowledge regarding inflammation and review the roles of intracellular inflammatory signaling pathways, which are specific inflammatory mediators in PD. Finally, possible therapeutic strategies are proposed, which may downregulate inflammatory processes and inhibit the progression of PD.
Collapse
Affiliation(s)
- Junqiang Yan
- Department of Neurology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Qizhi Fu
- Department of Neurology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Liniu Cheng
- Department of Neurology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Mingming Zhai
- Department of Neurology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Wenjuan Wu
- Department of Neurology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Lina Huang
- Department of Neurology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Ganqin Du
- Department of Neurology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| |
Collapse
|
23
|
Li MD, Burns TC, Morgan AA, Khatri P. Integrated multi-cohort transcriptional meta-analysis of neurodegenerative diseases. Acta Neuropathol Commun 2014; 2:93. [PMID: 25187168 PMCID: PMC4167139 DOI: 10.1186/s40478-014-0093-y] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 07/23/2014] [Indexed: 01/11/2023] Open
Abstract
Introduction Neurodegenerative diseases share common pathologic features including neuroinflammation, mitochondrial dysfunction and protein aggregation, suggesting common underlying mechanisms of neurodegeneration. We undertook a meta-analysis of public gene expression data for neurodegenerative diseases to identify a common transcriptional signature of neurodegeneration. Results Using 1,270 post-mortem central nervous system tissue samples from 13 patient cohorts covering four neurodegenerative diseases, we identified 243 differentially expressed genes, which were similarly dysregulated in 15 additional patient cohorts of 205 samples including seven neurodegenerative diseases. This gene signature correlated with histologic disease severity. Metallothioneins featured prominently among differentially expressed genes, and functional pathway analysis identified specific convergent themes of dysregulation. MetaCore network analyses revealed various novel candidate hub genes (e.g. STAU2). Genes associated with M1-polarized macrophages and reactive astrocytes were strongly enriched in the meta-analysis data. Evaluation of genes enriched in neurons revealed 70 down-regulated genes, over half not previously associated with neurodegeneration. Comparison with aging brain data (3 patient cohorts, 221 samples) revealed 53 of these to be unique to neurodegenerative disease, many of which are strong candidates to be important in neuropathogenesis (e.g. NDN, NAP1L2). ENCODE ChIP-seq analysis predicted common upstream transcriptional regulators not associated with normal aging (REST, RBBP5, SIN3A, SP2, YY1, ZNF143, IKZF1). Finally, we removed genes common to neurodegeneration from disease-specific gene signatures, revealing uniquely robust immune response and JAK-STAT signaling in amyotrophic lateral sclerosis. Conclusions Our results implicate pervasive bioenergetic deficits, M1-type microglial activation and gliosis as unifying themes of neurodegeneration, and identify numerous novel genes associated with neurodegenerative processes. Electronic supplementary material The online version of this article (doi:10.1186/s40478-014-0093-y) contains supplementary material, which is available to authorized users.
Collapse
|
24
|
Duan X, Tong J, Xu Q, Wu Y, Cai F, Li T, Song W. Upregulation of human PINK1 gene expression by NFκB signalling. Mol Brain 2014; 7:57. [PMID: 25108683 PMCID: PMC4237968 DOI: 10.1186/s13041-014-0057-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 07/29/2014] [Indexed: 11/26/2022] Open
Abstract
Parkinson’s disease (PD) is one of the major neurodegenerative disorders. Mitochondrial malfunction is implicated in PD pathogenesis. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN)-induced putative kinase 1 (PINK1), a serine/threonine kinase, plays an important role in the quality control of mitochondria and more than 70 PINK1 mutations have been identified to cause early-onset PD. However, the regulation of PINK1 gene expression remains elusive. In the present study, we identified the transcription start site (TSS) of the human PINK1 gene using switching mechanism at 5’end of RNA transcription (SMART RACE) assay. The TSS is located at 91 bp upstream of the translation start site ATG. The region with 104 bp was identified as the minimal promoter region by deletion analysis followed by dual luciferase assay. Four functional cis-acting nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB)-binding sites within the PINK1 promoter were identified. NFκB overexpression led to the up-regulation of PINK1 expression in both HEK293 cells and SH-SY5Y cells. Consistently, lipopolysaccharide (LPS), a strong activator of NFκB, significantly increased PINK1 expression in SH-SY5Y cells. Taken together, our results clearly suggested that PINK1 expression is tightly regulated at its transcription level and NFκB is a positive regulator for PINK1 expression.
Collapse
|
25
|
PAR2-mediated epigenetic upregulation of α-synuclein contributes to the pathogenesis of Parkinson׳s disease. Brain Res 2014; 1565:82-9. [DOI: 10.1016/j.brainres.2014.04.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 04/10/2014] [Accepted: 04/11/2014] [Indexed: 12/22/2022]
|