1
|
Kazmi S, Salehi-Pourmehr H, Sadigh-Eteghad S, Farhoudi M. The efficacy and safety of interleukin-1 receptor antagonist in stroke patients: A systematic review. J Clin Neurosci 2024; 120:120-128. [PMID: 38237490 DOI: 10.1016/j.jocn.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/29/2023] [Accepted: 01/08/2024] [Indexed: 02/12/2024]
Abstract
Stroke is the leading cause of disability worldwide, yet there is currently no effective treatment available to mitigate its negative consequences. Pro-inflammatory cytokines, such as interleukin-1 (IL-1), are known to play a crucial role in exacerbating the aftermath of stroke. Thus, it is hypothesized that blocking inflammation and administering anti-inflammatory drugs at an optimal time and dosage may improve the long-term quality of life for stroke patients. This systematic review examines the effectiveness and safety of IL-1 receptor antagonist (IL-1Ra), commercially known as "anakinra," in clinical studies involving the treatment of stroke patients. A comprehensive literature search was conducted until October 2023 to identify relevant studies. The search yielded 1403 articles, out of which 598 were removed due to duplication. After a thorough review of 805 titles and abstracts, 797 articles were further excluded, resulting in 8 studies being included in this systematic review. The findings from all the included studies demonstrate that IL-1Ra is safe for use in acute ischemic and hemorrhagic stroke patients, with no significant adverse events reported. Additionally, biomarkers, clinical assessments, serious adverse events (AEs), and non-serious AEs consistently showed more favorable outcomes in IL-1Ra receiving patients. Stroke elevates the levels of several inflammatory cytokines, however, administration of IL-1RA directly or indirectly modulates these markers and improves some clinical outcomes, suggesting a potential therapeutic benefit of this intervention.
Collapse
Affiliation(s)
- Sareh Kazmi
- Department of Neuroscience, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Neurosciences Research Center (NSRC), Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hanieh Salehi-Pourmehr
- Research Center for Evidence-Base Medicine, Iranian EBM Centre: A JBI Centre of Excellence, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Medical Philosophy and History Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center (NSRC), Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mehdi Farhoudi
- Neurosciences Research Center (NSRC), Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
2
|
Chen Y, Yang L, Wang C, Wang C. Exploring the mechanism of resistance to vincristine in breast cancer cells using transcriptome sequencing technology. Oncol Lett 2023; 26:502. [PMID: 37920438 PMCID: PMC10618930 DOI: 10.3892/ol.2023.14089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 09/21/2023] [Indexed: 11/04/2023] Open
Abstract
Breast cancer has replaced lung cancer as the leading cancer globally, but various chemotherapy drugs for breast cancer are prone to resistance, especially in patients with distant metastases who are susceptible to multiple chemotherapy drug resistance often leading to treatment failure. Vincristine (VCR) is an alkaloid extracted from Catharanthus roseus, and is often used in combination with other chemotherapy drugs to treat various types of cancer, including breast cancer. Research on the development of resistance to VCR has been carried out using transcriptome sequencing technology. Firstly, gradient increase of VCR concentration was used to produce a VCR-resistant breast cancer cell line. Mechanistically, RNA was extracted from the VCR-resistant breast cancer cell line, and the transcriptome was sequenced. Further analysis showed changes in the expression levels of various genes in the aforementioned VCR-resistant breast cancer cell line. Meanwhile, the analysis of splicing events also indicated a change in variable splicing events. Further validation showed that the expression levels of multiple genes, including interleukin-1β, were altered in the VCR-resistant breast cancer cell line, and these gene expression changes were related to VCR resistance. The results of the present study provide a theoretical basis for exploring the mechanism of VCR resistance clinically.
Collapse
Affiliation(s)
- Yao Chen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Lili Yang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Chao Wang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Changmiao Wang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| |
Collapse
|
3
|
Ma B, Liu X, Zhang Z, Ma C, Chand R, Patwardhan S, Wang C, Thamphiwatana SD, Chen P, Chen W. A digital nanoplasmonic microarray immunosensor for multiplexed cytokine monitoring during CAR T-cell therapy from a leukemia tumor microenvironment model. Biosens Bioelectron 2023; 230:115247. [PMID: 37023552 PMCID: PMC10103176 DOI: 10.1016/j.bios.2023.115247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 03/28/2023]
Abstract
The release of cytokines by chimeric antigen receptor (CAR) T-cells and tumor resident immune cells defines a significant part of CAR T-cell functional activity and patient immune responses during CAR T-cell therapy. However, few studies have so far precisely characterized the cytokine secretion dynamics in the tumor niche during CAR T-cell therapy, which requires multiplexed, and timely biosensing platforms and integration with biomimetic tumor microenvironment. Herein, we implemented a digital nanoplasmonic microarray immunosensor with a microfluidic biomimetic Leukemia-on-a-Chip model to monitor cytokine secretion dynamics during CD19 CAR T-cell therapy against precursor B-cell acute lymphocytic leukemia (B-ALL). The integrated nanoplasmonic biosensors achieved precise multiplexed cytokine measurements with low operating sample volume, short assay time, heightened sensitivity, and negligible sensor crosstalk. Using the digital nanoplasmonic biosensing approach, we measured the concentrations of six cytokines (TNF-α, IFN-γ, MCP-1, GM-CSF, IL-1β, and IL-6) during first 5 days of CAR T-cell treatment in the microfluidic Leukemia-on-a-Chip model. Our results revealed a heterogeneous secretion profile of various cytokines during CAR T-cell therapy and confirmed a correlation between the cytokine secretion profile and the CAR T-cell cytotoxic activity. The capability to monitor immune cell cytokine secretion dynamics in a biomimetic tumor microenvironment could further help in study of cytokine release syndrome during CAR T-cell therapy and in development of more efficient and safer immunotherapies.
Collapse
Affiliation(s)
- Benteng Ma
- Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Xinya Liu
- Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Zhuoyu Zhang
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Chao Ma
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Rashik Chand
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Saee Patwardhan
- Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Chuanyu Wang
- Department of Material Engineering, Auburn University, Auburn, AL, 36849, USA
| | - Soracha D Thamphiwatana
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakorn Pathom, 73170, Thailand
| | - Pengyu Chen
- Department of Material Engineering, Auburn University, Auburn, AL, 36849, USA
| | - Weiqiang Chen
- Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA; Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA; Perlmutter Cancer Center, NYU Langone Health, New York, NY, 10016, USA.
| |
Collapse
|
4
|
Ramos-Guzmán CA, Ruiz-Pernía JJ, Zinovjev K, Tuñón I. Unveiling the Mechanistic Singularities of Caspases: A Computational Analysis of the Reaction Mechanism in Human Caspase-1. ACS Catal 2023; 13:4348-4361. [PMID: 37066044 PMCID: PMC10088814 DOI: 10.1021/acscatal.3c00037] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/27/2023] [Indexed: 03/17/2023]
Abstract
Caspases are cysteine proteases in charge of breaking a peptide bond next to an aspartate residue. Caspases constitute an important family of enzymes involved in cell death and inflammatory processes. A plethora of diseases, including neurological and metabolic diseases and cancer, are associated with the poor regulation of caspase-mediated cell death and inflammation. Human caspase-1 in particular carries out the transformation of the pro-inflammatory cytokine pro-interleukin-1β into its active form, a key process in the inflammatory response and then in many diseases, such as Alzheimer's disease. Despite its importance, the reaction mechanism of caspases has remained elusive. The standard mechanistic proposal valid for other cysteine proteases and that involves the formation of an ion pair in the catalytic dyad is not supported by experimental evidence. Using a combination of classical and hybrid DFT/MM simulations, we propose a reaction mechanism for the human caspase-1 that explains experimental observations, including mutagenesis, kinetic, and structural data. In our mechanistic proposal, the catalytic cysteine, Cys285, is activated after a proton transfer to the amide group of the scissile peptide bond, a process facilitated by hydrogen-bond interactions with Ser339 and His237. The catalytic histidine does not directly participate in any proton transfer during the reaction. After formation of the acylenzyme intermediate, the deacylation step takes place through the activation of a water molecule by the terminal amino group of the peptide fragment formed during the acylation step. The overall activation free energy obtained from our DFT/MM simulations is in excellent agreement with the value derived from the experimental rate constant, 18.7 vs 17.9 kcal·mol-1, respectively. Simulations of the H237A mutant support our conclusions and agree with the reported reduced activity observed for this caspase-1 variant. We propose that this mechanism can explain the reactivity of all cysteine proteases belonging to the CD clan and that differences with respect to other clans could be related to the larger preference showed by enzymes of the CD clan for charged residues at position P1. This mechanism would avoid the free energy penalty associated with the formation of an ion pair. Finally, our structural description of the reaction process can be useful to assist in the design of inhibitors of caspase-1, a target in the treatment of several human diseases.
Collapse
Affiliation(s)
- Carlos A. Ramos-Guzmán
- Departamento de Química Física, Universitat de Valencia, 46100 Burjassot, Spain
- Instituto de Materiales Avanzados, Universitat Jaume I, 12071 Castelló, Spain
| | | | - Kirill Zinovjev
- Departamento de Química Física, Universitat de Valencia, 46100 Burjassot, Spain
| | - Iñaki Tuñón
- Departamento de Química Física, Universitat de Valencia, 46100 Burjassot, Spain
| |
Collapse
|
5
|
Derivation and Validation of a New Visceral Adiposity Index for Predicting Short-Term Mortality of Patients with Acute Ischemic Stroke in a Chinese Population. Brain Sci 2023; 13:brainsci13020297. [PMID: 36831840 PMCID: PMC9954352 DOI: 10.3390/brainsci13020297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/20/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
The visceral adiposity index (VAI) is related to the occurrence of various cardiometabolic diseases, atherosclerosis, and stroke. However, few studies have analyzed the impact on the short-term prognosis of stroke. We assessed the effect of VAI on short-term prognoses in patients with acute ischemic stroke through a retrospective cohort study of 225 patients with acute stroke who were admitted to the neurological intensive care unit of our hospital. We collected metabolic indicators (blood pressure, fasting glucose, lipids), National Institutes of Health Stroke Scale (NIHSS) scores, symptomatic intracranial hemorrhage, and other disease evaluation indicators on 197 patients who were screened for inclusion. VAI was calculated by using baseline data (sex, height, weight, waist circumference (WC)). We assessed functional recovery according to modified Rankin scale scores after 90 days. The receiver operating characteristic (ROC) curve was used to calculate the VAI cutoff value that affects short-term outcomes. A nomogram that can predict the risk of short-term mortality in patients with acute ischemic stroke was drawn. In total, 28 patients died within 90 days. Those patients had higher VAI (p = 0.000), higher triglyceride (TG) (p = 0.020) and NIHSS scores (p = 0.000), and lower high-density lipoprotein cholesterol (HDL-C) (p = 0.000) than patients who survived. VAI had higher predictive value of short-term mortality than did body mass index (BMI), body fat mass index (BFMI), and WC. VAI and NIHSS scores were independent risk factors for the short-term mortality of patients with stroke. Patients with a VAI > 2.355 had a higher risk of short-term mortality. VAI has a predictive value higher than that of traditional metabolic indicators such as BMI, BFMI, and WC. The nomogram, composed of NIHSS, VAI, HDL-C, and TG, may predict the short-term mortality of cerebral infarction patients.
Collapse
|
6
|
Pandey V, Shukla D, Nirmal S, Devi BI, Christopher R. Biomarkers in Traumatic Brain Injuries: Narrative Review. INDIAN JOURNAL OF NEUROTRAUMA 2022. [DOI: 10.1055/s-0042-1759853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
AbstractTraumatic brain injury (TBI) is a multistep interaction of brain antigens, cytokine-mediated humeral, and cellular immune reactions. Because of the limitations of clinical and radiological evaluation in TBI, there has been a considerable advancement toward the need for developing biomarkers that can predict the severity of TBI. Blood-based brain biomarkers hold the potential to predict the absence of intracranial injury and thus decrease unnecessary brain computed tomographic scanning. Various biomarkers have been studied that detects neuronal, axonal, and blood–brain barrier integrity. Biomarkers are still under investigation and hold promise in the future evaluation of TBI patients. They can be used for grading as well as a prognostication of head injury.
Collapse
Affiliation(s)
- Vishram Pandey
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, NIMHANS, Bangalore, Karnataka, India
| | - Dhaval Shukla
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, NIMHANS, Bangalore, Karnataka, India
| | - Shubham Nirmal
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, NIMHANS, Bangalore, Karnataka, India
| | - Bhagavatula Indira Devi
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, NIMHANS, Bangalore, Karnataka, India
| | - Rita Christopher
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, NIMHANS, Bangalore, Karnataka, India
| |
Collapse
|
7
|
Zhu H, Hu S, Li Y, Sun Y, Xiong X, Hu X, Chen J, Qiu S. Interleukins and Ischemic Stroke. Front Immunol 2022; 13:828447. [PMID: 35173738 PMCID: PMC8841354 DOI: 10.3389/fimmu.2022.828447] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/12/2022] [Indexed: 12/17/2022] Open
Abstract
Ischemic stroke after cerebral artery occlusion is one of the major causes of chronic disability worldwide. Interleukins (ILs) play a bidirectional role in ischemic stroke through information transmission, activation and regulation of immune cells, mediating the activation, multiplication and differentiation of T and B cells and in the inflammatory reaction. Crosstalk between different ILs in different immune cells also impact the outcome of ischemic stroke. This overview is aimed to roughly discuss the multiple roles of ILs after ischemic stroke. The roles of IL-1, IL-2, IL-4, IL-5, IL-6, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, IL-16, IL-17, IL-18, IL-19, IL-21, IL-22, IL-23, IL-32, IL-33, IL-34, IL-37, and IL-38 in ischemic stroke were discussed in this review.
Collapse
Affiliation(s)
- Hua Zhu
- Department of Neurosurgery, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), Huzhou, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Siping Hu
- Department of Anesthesiology, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), Huzhou, China
| | - Yuntao Li
- Department of Neurosurgery, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), Huzhou, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yao Sun
- Department of Neurosurgery, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), Huzhou, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), Huzhou, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinyao Hu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Junjing Chen
- Department of General Surgery, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), Huzhou, China
- *Correspondence: Junjing Chen, ; Sheng Qiu,
| | - Sheng Qiu
- Department of Neurosurgery, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), Huzhou, China
- *Correspondence: Junjing Chen, ; Sheng Qiu,
| |
Collapse
|
8
|
Wong LM, Phoon LQ, Wei LK. Epigenetics Modifications in Large-Artery Atherosclerosis: A Systematic Review. J Stroke Cerebrovasc Dis 2021; 30:106033. [PMID: 34598837 DOI: 10.1016/j.jstrokecerebrovasdis.2021.106033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 07/15/2021] [Accepted: 08/01/2021] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVES In recent years, the evidence of the relationship between epigenetics and acute ischemic stroke (AIS) were accumulating, however, the epigenetic characteristics that directs specifically towards the aetiology of large-artery atherosclerosis (LAA) remain ambiguous. The aim of this study was to highlight the overall evidence concerning the epigenetic mechanisms associated with the occurrence of LAA. MATERIALS AND METHODS Studies that involve investigations related to epigenetic markers (DNA methylation and RNA modifications) and LAA were retrieved from eleven scientific publication databases. The studies were screened through the pre-set inclusion and exclusion criteria prior to the NOS evaluation. RESULTS Eligible studies (n=25) were evaluated. Of which, six reported on DNA methylation and 19 studies assessed RNA modifications (16 on miRNAs, two on lncRNAs, and one study on circRNA). Hypomethylation of MTRNR2L8 and ERα promoters; microRNAs (miR-7-2-3p, miR-16, miR-34a-5p, miR-126, miR-143, miR-200b, miR-223, miR-503, miR-1908, miR-146a rs2910164 C/G, miR-149 rs2292832 T/C, miR-200b rs7549819 T/C, miR-34a rs2666433); lncRNA of ZFAS1; and circRNA of hsa_circRNA_102488 were associated with LAA significantly. CONCLUSION Current systematic review highlighted hypomethylation of miRNAs and lncRNA might be the potential biomarkers for LAA.
Collapse
Affiliation(s)
- Li Min Wong
- Department of Biological Science, Faculty of Science, Universiti Tunku Abdul Rahman, Bandar Barat, Kampar, Perak 31900, Malaysia
| | - Lee Quen Phoon
- Department of Allied Health Sciences, Faculty of Science, Universiti Tunku Abdul Rahman, Bandar Barat, Kampar, Perak 31900, Malaysia
| | - Loo Keat Wei
- Department of Biological Science, Faculty of Science, Universiti Tunku Abdul Rahman, Bandar Barat, Kampar, Perak 31900, Malaysia.
| |
Collapse
|
9
|
Harati-Sadegh M, Mohammadoo-Khorasani M, Sargazi S, Saravani R, Shahraki S, Eskandari E. Quantitative Assessment of the Effects of IL-1ß -511 C>T Variant on Breast Cancer Risk: An Updated Meta-Analysis of 3331 Cases and 3609 Controls. Lab Med 2021; 52:36-46. [PMID: 32754752 DOI: 10.1093/labmed/lmaa055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
OBJECTIVE Growing evidence suggests that IL-1β -511C>T, as a functional variant, affects the risk of developing breast cancer (BC); however, the results have not been conclusive. This meta-analysis was conducted to estimate the link between this variant and BC risk. METHODS We retrieved available publications on IL-1β -511C>T polymorphism by conducting a comprehensive literature search on the Web of Science, MEDLINE, PubMed, Scopus, and Google scholar databases (last search on February 25, 2020). RESULTS The overall analysis indicates that IL-1β -511C>T polymorphism conferred an increased risk of BC under a recessive TT vs CT+CC model by 1.14-fold and showed protection against BC under an overdominant CT vs TT+CC genetic contrast model (odds ratio = 0.84). Stratified analysis based on ethnicity revealed the protective effect of this single-nucleotide polymorphism against BC risk in Caucasian patients. CONCLUSION Our data results provide a proof of concept for the association of IL-1β -511C>T with BC risk. Larger, well-designed population-based studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Mahdiyeh Harati-Sadegh
- Genetics of Non-Communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Milad Mohammadoo-Khorasani
- Department of Clinical Biochemistry, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Saman Sargazi
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute
| | - Ramin Saravani
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute.,Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Sheida Shahraki
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute
| | - Ebrahim Eskandari
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
10
|
Garcia-Bonilla L, Sciortino R, Shahanoor Z, Racchumi G, Janakiraman M, Montaner J, Zhou P, Anrather J, Iadecola C. Role of microglial and endothelial CD36 in post-ischemic inflammasome activation and interleukin-1β-induced endothelial activation. Brain Behav Immun 2021; 95:489-501. [PMID: 33872708 PMCID: PMC8187325 DOI: 10.1016/j.bbi.2021.04.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 04/08/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
Cerebral ischemia is associated with an acute inflammatory response that contributes to the resulting injury. The innate immunity receptor CD36, expressed in microglia and endothelium, and the pro-inflammatory cytokine interleukin-1β (IL-1β) are involved in the mechanisms of ischemic injury. Since CD36 has been implicated in activation of the inflammasome, the main source of IL-1β, we investigated whether CD36 mediates brain injury through the inflammasome and IL-1β. We found that active caspase-1, a key inflammasome component, is decreased in microglia of CD36-deficient mice subjected to transient middle cerebral artery occlusion, an effect associated with a reduction in brain IL-1β. Conditional deletion of CD36 either in microglia or endothelium reduced ischemic injury in mice, attesting to the pathogenic involvement of CD36 in both cell types. Application of an ischemic brain extract to primary brain endothelial cell cultures from wild type (WT) mice induced IL-1β-dependent endothelial activation, reflected by increases in the cytokine colony stimulating factor-3, a response markedly attenuated in CD36-deficient endothelia. Similarly, the increase in colony stimulating factor-3 induced by recombinant IL-1β was attenuated in CD36-deficient compared to WT endothelia. We conclude that microglial CD36 is a key determinant of post-ischemic IL-1β production by regulating caspase-1 activity, whereas endothelial CD36 is required for the full expression of the endothelial activation induced by IL-1β. The data identify microglial and endothelial CD36 as critical upstream components of the acute inflammatory response to cerebral ischemia and viable putative therapeutic targets.
Collapse
Affiliation(s)
- Lidia Garcia-Bonilla
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA.
| | - Rose Sciortino
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ziasmin Shahanoor
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Gianfranco Racchumi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Mathangi Janakiraman
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Joan Montaner
- Neurovascular Lab, Vall d́Hebron Research Institute (VHIR), 08035 Barcelona, Spain
| | - Ping Zhou
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA.
| |
Collapse
|
11
|
D’Amico R, Trovato Salinaro A, Fusco R, Cordaro M, Impellizzeri D, Scuto M, Ontario ML, Lo Dico G, Cuzzocrea S, Di Paola R, Siracusa R, Calabrese V. Hericium erinaceus and Coriolus versicolor Modulate Molecular and Biochemical Changes after Traumatic Brain Injury. Antioxidants (Basel) 2021; 10:898. [PMID: 34199629 PMCID: PMC8228340 DOI: 10.3390/antiox10060898] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/21/2021] [Accepted: 05/29/2021] [Indexed: 01/30/2023] Open
Abstract
Traumatic brain injury (TBI) is a major health and socioeconomic problem affecting the world. This condition results from the application of external physical force to the brain which leads to transient or permanent structural and functional impairments. TBI has been shown to be a risk factor for neurodegeneration which can lead to Parkinson's disease (PD) for example. In this study, we wanted to explore the development of PD-related pathology in the context of an experimental model of TBI and the potential ability of Coriolus versicolor and Hericium erinaceus to prevent neurodegenerative processes. Traumatic brain injury was induced in mice by controlled cortical impact. Behavioral tests were performed at various times: the animals were sacrificed 30 days after the impact and the brain was processed for Western blot and immunohistochemical analyzes. After the head injury, a significant decrease in the expression of tyrosine hydroxylase and the dopamine transporter in the substantia nigra was observed, as well as significant behavioral alterations that were instead restored following daily oral treatment with Hericium erinaceus and Coriolus versicolor. Furthermore, a strong increase in neuroinflammation and oxidative stress emerged in the vehicle groups. Treatment with Hericium erinaceus and Coriolus versicolor was able to prevent both the neuroinflammatory and oxidative processes typical of PD. This study suggests that PD-related molecular events may be triggered on TBI and that nutritional fungi such as Hericium erinaceus and Coriolus versicolor may be important in redox stress response mechanisms and neuroprotection, preventing the progression of neurodegenerative diseases such as PD.
Collapse
Affiliation(s)
- Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.D.); (R.F.); (D.I.); (R.S.)
| | - Angela Trovato Salinaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy; (A.T.S.); (M.S.); (M.L.O.); (G.L.D.); (V.C.)
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.D.); (R.F.); (D.I.); (R.S.)
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.D.); (R.F.); (D.I.); (R.S.)
| | - Maria Scuto
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy; (A.T.S.); (M.S.); (M.L.O.); (G.L.D.); (V.C.)
| | - Maria Laura Ontario
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy; (A.T.S.); (M.S.); (M.L.O.); (G.L.D.); (V.C.)
| | - Gianluigi Lo Dico
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy; (A.T.S.); (M.S.); (M.L.O.); (G.L.D.); (V.C.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.D.); (R.F.); (D.I.); (R.S.)
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA
| | - Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.D.); (R.F.); (D.I.); (R.S.)
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.D.); (R.F.); (D.I.); (R.S.)
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy; (A.T.S.); (M.S.); (M.L.O.); (G.L.D.); (V.C.)
| |
Collapse
|
12
|
Fourie C, Shridas P, Davis T, de Villiers WJ, Engelbrecht AM. Serum amyloid A and inflammasome activation: A link to breast cancer progression? Cytokine Growth Factor Rev 2021; 59:62-70. [DOI: 10.1016/j.cytogfr.2020.10.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022]
|
13
|
Tuo QZ, Zhang ST, Lei P. Mechanisms of neuronal cell death in ischemic stroke and their therapeutic implications. Med Res Rev 2021; 42:259-305. [PMID: 33957000 DOI: 10.1002/med.21817] [Citation(s) in RCA: 280] [Impact Index Per Article: 93.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 03/31/2021] [Accepted: 04/23/2021] [Indexed: 02/05/2023]
Abstract
Ischemic stroke caused by arterial occlusion is the most common type of stroke, which is among the most frequent causes of disability and death worldwide. Current treatment approaches involve achieving rapid reperfusion either pharmacologically or surgically, both of which are time-sensitive; moreover, blood flow recanalization often causes ischemia/reperfusion injury. However, even though neuroprotective intervention is urgently needed in the event of stroke, the exact mechanisms of neuronal death during ischemic stroke are still unclear, and consequently, the capacity for drug development has remained limited. Multiple cell death pathways are implicated in the pathogenesis of ischemic stroke. Here, we have reviewed these potential neuronal death pathways, including intrinsic and extrinsic apoptosis, necroptosis, autophagy, ferroptosis, parthanatos, phagoptosis, and pyroptosis. We have also reviewed the latest results of pharmacological studies on ischemic stroke and summarized emerging drug targets with a focus on clinical trials. These observations may help to further understand the pathological events in ischemic stroke and bridge the gap between basic and translational research to reveal novel neuroprotective interventions.
Collapse
Affiliation(s)
- Qing-Zhang Tuo
- Department of Geriatrics and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Shu-Ting Zhang
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
14
|
Blume C, Geiger MF, Müller M, Clusmann H, Mainz V, Kalder J, Brandenburg LO, Mueller CA. Decreased angiogenesis as a possible pathomechanism in cervical degenerative myelopathy. Sci Rep 2021; 11:2497. [PMID: 33510227 PMCID: PMC7843718 DOI: 10.1038/s41598-021-81766-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 01/07/2021] [Indexed: 01/03/2023] Open
Abstract
Endogenous immune mediated reactions of inflammation and angiogenesis are components of the spinal cord injury in patients with degenerative cervical myelopathy (DCM). The aim of this study was to identify alteration of certain mediators participating in angiogenetic and inflammatory reactions in patients with DCM. A consecutive series of 42 patients with DCM and indication for surgical decompression were enrolled for the study. 28 DCM patients were included, as CSF samples were taken preoperatively. We enrolled 42 patients requiring surgery for a thoracic abdominal aortic aneurysm (TAAA) as neurologically healthy controls. In 38 TAAA patients, CSF samples were taken prior to surgery and thus included. We evaluated the neurological status of patients and controls prior to surgery including NDI and mJOA. Protein-concentrations of factors with a crucial role in inflammation and angiogenesis were measured in CSF via ELISA testing (pg/ml): Angiopoietin 2, VEGF-A and C, RANTES, IL 1 beta and IL 8. Additionally, evaluated the status of the blood-spinal cord barrier (BSCB) by Reibers´diagnostic in all participants. Groups evidently differed in their neurological status (mJOA: DCM 10.1 ± 3.3, TAAA 17.3 ± 1.2, p < .001; NDI: DCM 47.4 ± 19.7, TAAA 5.3 ± 8.6, p < .001). There were no particular differences in age and gender distribution. However, we detected statistically significant differences in concentrations of mediators between the groups: Angiopoietin 2 (DCM 267.1.4 ± 81.9, TAAA 408.6 ± 177.1, p < .001) and VEGF C (DCM 152.2 ± 96.1, TAAA 222.4 ± 140.3, p = .04). DCM patients presented a mild to moderate BSCB disruption, controls had no signs of impairment. In patients with DCM, we measured decreased concentrations of angiogenic mediators. These results correspond to findings of immune mediated secondary harm in acute spinal cord injury. Reduced angiogenic activity could be a relevant part of the pathogenesis of DCM and secondary harm to the spinal cord.
Collapse
Affiliation(s)
- Christian Blume
- Department of Neurosurgery, RWTH Aachen University, Pauwelstrasse 30, 52074, Aachen, Germany.
| | - M F Geiger
- Department of Neurosurgery, RWTH Aachen University, Pauwelstrasse 30, 52074, Aachen, Germany
| | - M Müller
- Department of Neuroradiology, RWTH Aachen University, Pauwelstrasse 30, 52074, Aachen, Germany
| | - H Clusmann
- Department of Neurosurgery, RWTH Aachen University, Pauwelstrasse 30, 52074, Aachen, Germany
| | - V Mainz
- Department of Medical Psychology and Medical Sociology, RWTH Aachen University, Pauwelsstrasse 19, 52074, Aachen, Germany
| | - J Kalder
- Department of Vascular Surgery, Gießen University, Rudolf-Buchheim-str. 7, 35392, Gießen, Germany
| | - L O Brandenburg
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, 18057, Rostock, Germany
| | - C A Mueller
- Department of Neurosurgery, RWTH Aachen University, Pauwelstrasse 30, 52074, Aachen, Germany
| |
Collapse
|
15
|
Hernandez-Ronquillo L, Miranzadeh Mahabadi H, Moien-Afshari F, Wu A, Auer R, Zherebitskiy V, Borowsky R, Mickleborough M, Huntsman R, Vrbancic M, Cayabyab FS, Taghibiglou C, Carter A, Tellez-Zenteno JF. The Concept of an Epilepsy Brain Bank. Front Neurol 2020; 11:833. [PMID: 32973652 PMCID: PMC7468480 DOI: 10.3389/fneur.2020.00833] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/03/2020] [Indexed: 12/31/2022] Open
Abstract
Epilepsy comprises more than 40 clinical syndromes affecting millions of patients and families worldwide. To decode the molecular and pathological framework of epilepsy researchers, need reliable human epilepsy and control brain samples. Brain bank organizations collecting and supplying well-documented clinically and pathophysiologically tissue specimens are important for high-quality neurophysiology and neuropharmacology studies for epilepsy and other neurological diseases. New development in molecular mechanism and new treatment methods for neurological disorders have evoked increased demands for human brain tissue. An epilepsy brain bank is a storage source for both the frozen samples as well as the formaldehyde fixed paraffin embedded (FFPE) tissue from epilepsy surgery resections. In 2014, the University of Saskatchewan have started collecting human epilepsy brain tissues for the first time in Canada. This review highlights the necessity and importance of Epilepsy Brain bank that provides unique access for research to valuable source of brain tissue and blood samples from epilepsy patients.
Collapse
Affiliation(s)
- Lizbeth Hernandez-Ronquillo
- Saskatchewan Epilepsy Program, Division of Neurology, Department of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Hajar Miranzadeh Mahabadi
- Department of Anatomy, Physiology and Pharmacology, College of Medicine University of Saskatchewan, Saskatoon, SK, Canada
| | | | - Adam Wu
- Division of Neurosurgery, Department of Surgery, University of Saskatchewan, Saskatoon, SK, Canada
| | - Roland Auer
- Department of Pathology and Laboratory Medicine, Royal University Hospital, Saskatchewan Health Region, University of Saskatchewan, Saskatoon, SK, Canada
| | - Viktor Zherebitskiy
- Department of Pathology and Laboratory Medicine, Royal University Hospital, Saskatchewan Health Region, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ron Borowsky
- Cognitive Neuroscience Laboratory, Department of Psychology, College of Arts and Science, University of Saskatchewan, Saskatoon, SK, Canada
| | - Marla Mickleborough
- Cognitive Neuroscience Laboratory, Department of Psychology, College of Arts and Science, University of Saskatchewan, Saskatoon, SK, Canada
| | - Richard Huntsman
- Division of Pediatric Neurology, Department of Pediatrics, University of Saskatchewan, Saskatoon, SK, Canada
| | - Mirna Vrbancic
- Department of Clinical Health Psychology, Ellis Hall, Royal University Hospital, Saskatoon, SK, Canada
| | - Francisco S Cayabyab
- Division of Neurosurgery, Department of Surgery, University of Saskatchewan, Saskatoon, SK, Canada
| | - Changiz Taghibiglou
- Department of Anatomy, Physiology and Pharmacology, College of Medicine University of Saskatchewan, Saskatoon, SK, Canada
| | - Alexandra Carter
- Saskatchewan Epilepsy Program, Division of Neurology, Department of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Jose F Tellez-Zenteno
- Saskatchewan Epilepsy Program, Division of Neurology, Department of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
16
|
Yan W, Sun W, Fan J, Wang H, Han S, Li J, Yin Y. Sirt1-ROS-TRAF6 Signaling-Induced Pyroptosis Contributes to Early Injury in Ischemic Mice. Neurosci Bull 2020; 36:845-859. [PMID: 32253651 PMCID: PMC7410906 DOI: 10.1007/s12264-020-00489-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 10/04/2019] [Indexed: 01/01/2023] Open
Abstract
Stroke is an acute cerebro-vascular disease with high incidence and poor prognosis, most commonly ischemic in nature. In recent years, increasing attention has been paid to inflammatory reactions as symptoms of a stroke. However, the role of inflammation in stroke and its underlying mechanisms require exploration. In this study, we evaluated the inflammatory reactions induced by acute ischemia and found that pyroptosis occurred after acute ischemia both in vivo and in vitro, as determined by interleukin-1β, apoptosis-associated speck-like protein, and caspase-1. The early inflammation resulted in irreversible ischemic injury, indicating that it deserves thorough investigation. Meanwhile, acute ischemia decreased the Sirtuin 1 (Sirt1) protein levels, and increased the TRAF6 (TNF receptor associated factor 6) protein and reactive oxygen species (ROS) levels. In further exploration, both Sirt1 suppression and TRAF6 activation were found to contribute to this pyroptosis. Reduced Sirt1 levels were responsible for the production of ROS and increased TRAF6 protein levels after ischemic exposure. Moreover, N-acetyl-L-cysteine, an ROS scavenger, suppressed the TRAF6 accumulation induced by oxygen-glucose deprivation via suppression of ROS bursts. These phenomena indicate that Sirt1 is upstream of ROS, and ROS bursts result in increased TRAF6 levels. Further, the activation of Sirt1 during the period of ischemia reduced ischemia-induced injury after 72 h of reperfusion in mice with middle cerebral artery occlusion. In sum, these results indicate that pyroptosis-dependent machinery contributes to the neural injury during acute ischemia via the Sirt1-ROS-TRAF6 signaling pathway. We propose that inflammatory reactions occur soon after oxidative stress and are detrimental to neuronal survival; this provides a promising therapeutic target against ischemic injuries such as a stroke.
Collapse
Affiliation(s)
- Weijie Yan
- Department of Neurobiology, Ministry of Education Key Laboratory for Neurodegenerative Disorders, Capital Medical University, Beijing, 100069, China
| | - Wei Sun
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Jiahui Fan
- Department of Neurobiology, Ministry of Education Key Laboratory for Neurodegenerative Disorders, Capital Medical University, Beijing, 100069, China
| | - Haiqing Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Song Han
- Department of Neurobiology, Ministry of Education Key Laboratory for Neurodegenerative Disorders, Capital Medical University, Beijing, 100069, China
| | - Junfa Li
- Department of Neurobiology, Ministry of Education Key Laboratory for Neurodegenerative Disorders, Capital Medical University, Beijing, 100069, China
| | - Yanling Yin
- Department of Neurobiology, Ministry of Education Key Laboratory for Neurodegenerative Disorders, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
17
|
Delic V, Beck KD, Pang KCH, Citron BA. Biological links between traumatic brain injury and Parkinson's disease. Acta Neuropathol Commun 2020; 8:45. [PMID: 32264976 PMCID: PMC7137235 DOI: 10.1186/s40478-020-00924-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 03/26/2020] [Indexed: 12/19/2022] Open
Abstract
Parkinson's Disease (PD) is a progressive neurodegenerative disorder with no cure. Clinical presentation is characterized by postural instability, resting tremors, and gait problems that result from progressive loss of A9 dopaminergic neurons in the substantia nigra pars compacta. Traumatic brain injury (TBI) has been implicated as a risk factor for several neurodegenerative diseases, but the strongest evidence is linked to development of PD. Mild TBI (mTBI), is the most common and is defined by minimal, if any, loss of consciousness and the absence of significant observable damage to the brain tissue. mTBI is responsible for a 56% higher risk of developing PD in U.S. Veterans and the risk increases with severity of injury. While the mounting evidence from human studies suggests a link between TBI and PD, fundamental questions as to whether TBI nucleates PD pathology or accelerates PD pathology in vulnerable populations remains unanswered. Several promising lines of research point to inflammation, metabolic dysregulation, and protein accumulation as potential mechanisms through which TBI can initiate or accelerate PD. Amyloid precursor protein (APP), alpha synuclein (α-syn), hyper-phosphorylated Tau, and TAR DNA-binding protein 43 (TDP-43), are some of the most frequently reported proteins upregulated following a TBI and are also closely linked to PD. Recently, upregulation of Leucine Rich Repeat Kinase 2 (LRRK2), has been found in the brain of mice following a TBI. Subset of Rab proteins were identified as biological substrates of LRRK2, a protein also extensively linked to late onset PD. Inhibition of LRRK2 was found to be neuroprotective in PD and TBI models. The goal of this review is to survey current literature concerning the mechanistic overlap between TBI and PD with a particular focus on inflammation, metabolic dysregulation, and aforementioned proteins. This review will also cover the application of rodent TBI models to further our understanding of the relationship between TBI and PD.
Collapse
Affiliation(s)
- Vedad Delic
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research and Development (Mailstop 15), 385 Tremont Ave, East Orange, NJ, 07018, USA.
- NeuroBehavioral Research Laboratory, VA New Jersey Health Care System, Research and Development (Mailstop 15), 385 Tremont Ave, East Orange, NJ, 07018, USA.
| | - Kevin D Beck
- NeuroBehavioral Research Laboratory, VA New Jersey Health Care System, Research and Development (Mailstop 15), 385 Tremont Ave, East Orange, NJ, 07018, USA
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers- New Jersey Medical School, Newark, NJ, 07103, USA
| | - Kevin C H Pang
- NeuroBehavioral Research Laboratory, VA New Jersey Health Care System, Research and Development (Mailstop 15), 385 Tremont Ave, East Orange, NJ, 07018, USA
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers- New Jersey Medical School, Newark, NJ, 07103, USA
| | - Bruce A Citron
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research and Development (Mailstop 15), 385 Tremont Ave, East Orange, NJ, 07018, USA
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers- New Jersey Medical School, Newark, NJ, 07103, USA
| |
Collapse
|
18
|
Liang J, Wang Q, Li JQ, Guo T, Yu D. Long non-coding RNA MEG3 promotes cerebral ischemia-reperfusion injury through increasing pyroptosis by targeting miR-485/AIM2 axis. Exp Neurol 2019; 325:113139. [PMID: 31794744 DOI: 10.1016/j.expneurol.2019.113139] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 11/28/2019] [Accepted: 11/29/2019] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Inflammasome contributes to ischemic brain injury by inducing pyroptosis and inflammation. The aim of this study is to unravel the mechanism of long non-coding RNA (lncRNA) maternally expressed gene 3 (MEG3)-mediated regulation of absent in melanoma 2 (AIM2) inflammasome during cerebral ischemia/reperfusion (I/R). METHODS In vivo middle cerebral artery occlusion (MCAO) rat model and in vitro oxygen-glucose deprivation/reperfusion (OGD/R)-treated neurocytes model were generated. TTC, H&E staining and TUNEL were performed to assess the cerebral ischemic injury. LDH and MTT assays were used to detect cell viability and cytotoxicity. qRT-PCR was used to detect the expression levels of MEG3, miR-485 and AIM2. Immunohistochemistry (IHC) and immunofluorescence were conducted to detect the AIM2 expression. ELISA and Western blotting were performed to determine the secretion and protein levels of inflammasome signaling proteins. Dual luciferase reporter assay and Ago2-RIP were used to validate the direct interaction among MEG3, miR-485 and AIM2. RESULTS In both MCAO rats and OGD/R-treated neurocytes, MEG3 and AIM2 were significantly up-regulated, whereas miR-485 was down-regulated. MCAO induces pyroptosis and release of IL-1β and IL-18 in ischemia brain. MEG3 acted as a molecular sponge to suppress miR-485, and AIM2 was identified as a direct target of miR-485. Knockdown of MEG3 inhibited OGD/R-induced pyroptosis and inflammation, and lack of MEG3 inhibited caspase1 signaling and decreased the expression of AIM2, ASC, cleaved-caspase1 and GSDMD-N. While overexpression of MEG3 exerted opposite effects. CONCLUSION MEG3/miR-485/AIM2 axis contributes to pyroptosis via activating caspase1 signaling during cerebral I/R, suggesting that this axis may be a potent therapeutic target in ischemic stroke.
Collapse
Affiliation(s)
- Ji Liang
- Department of Neurology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, Hainan Province, PR China
| | - Qiang Wang
- Department of Neurology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, Hainan Province, PR China
| | - Jun-Qi Li
- Department of Neurology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, Hainan Province, PR China
| | - Tie Guo
- Department of ICU, The First Affiliated Hospital of Zhengzhou University, PR China
| | - Dan Yu
- Department of Neurology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, Hainan Province, PR China.
| |
Collapse
|
19
|
Ahn SH, Savarraj JPJ, Parsha K, Hergenroeder GW, Chang TR, Kim DH, Kitagawa RS, Blackburn SL, Choi HA. Inflammation in delayed ischemia and functional outcomes after subarachnoid hemorrhage. J Neuroinflammation 2019; 16:213. [PMID: 31711504 PMCID: PMC6849179 DOI: 10.1186/s12974-019-1578-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/05/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Inflammatory mechanism has been implicated in delayed cerebral ischemia (DCI) and poor functional outcomes after subarachnoid hemorrhage (SAH). Identification of cytokine patterns associated with inflammation in acute SAH will provide insights into underlying biological processes of DCI and poor outcomes that may be amenable to interventions. METHODS Serum samples were collected from a prospective cohort of 60 patients with acute non-traumatic SAH at four time periods (< 24 h, 24-48 h, 3-5 days, and 6-8 days after SAH) and concentration levels of 41 cytokines were measured by multiplex immunoassay. Logistic regression analysis was used to identify cytokines associated with DCI and poor functional outcomes. Correlation networks were constructed to identify cytokine clusters. RESULTS Of the 60 patients enrolled in the study, 14 (23.3%) developed DCI and 16 (26.7%) had poor functional outcomes at 3 months. DCI was associated with increased levels of PDGF-ABBB and CCL5 and decreased levels of IP-10 and MIP-1α. Poor functional outcome was associated with increased levels of IL-6 and MCP-1α. Network analysis identified distinct cytokine clusters associated with DCI and functional outcomes. CONCLUSIONS Serum cytokine patterns in early SAH are associated with poor functional outcomes and DCI. The significant cytokines primarily modulate the inflammatory response. This supports earlier SAH studies linking inflammation and poor outcomes. In particular, this study identifies novel cytokine patterns over time that may indicate impending DCI.
Collapse
Affiliation(s)
- Sung-Ho Ahn
- Department of Neurology, Pusan National University School of Medicine, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Busan, South Korea
| | - Jude P J Savarraj
- Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, USA
| | - Kaushik Parsha
- Department of Neurology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Georgene W Hergenroeder
- Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, USA
| | - Tiffany R Chang
- Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, USA
| | - Dong H Kim
- Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, USA
| | - Ryan S Kitagawa
- Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, USA
| | - Spiros L Blackburn
- Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, USA
| | - H Alex Choi
- Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, USA.
| |
Collapse
|
20
|
Neuroinflammation in Post-Traumatic Epilepsy: Pathophysiology and Tractable Therapeutic Targets. Brain Sci 2019; 9:brainsci9110318. [PMID: 31717556 PMCID: PMC6895909 DOI: 10.3390/brainsci9110318] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 02/06/2023] Open
Abstract
Epilepsy is a common chronic consequence of traumatic brain injury (TBI), contributing to increased morbidity and mortality for survivors. As post-traumatic epilepsy (PTE) is drug-resistant in at least one-third of patients, there is a clear need for novel therapeutic strategies to prevent epilepsy from developing after TBI, or to mitigate its severity. It has long been recognized that seizure activity is associated with a local immune response, characterized by the activation of microglia and astrocytes and the release of a plethora of pro-inflammatory cytokines and chemokines. More recently, increasing evidence also supports a causal role for neuroinflammation in seizure induction and propagation, acting both directly and indirectly on neurons to promote regional hyperexcitability. In this narrative review, we focus on key aspects of the neuroinflammatory response that have been implicated in epilepsy, with a particular focus on PTE. The contributions of glial cells, blood-derived leukocytes, and the blood–brain barrier will be explored, as well as pro- and anti-inflammatory mediators. While the neuroinflammatory response to TBI appears to be largely pro-epileptogenic, further research is needed to clearly demonstrate causal relationships. This research has the potential to unveil new drug targets for PTE, and identify immune-based biomarkers for improved epilepsy prediction.
Collapse
|
21
|
Robicsek SA, Bhattacharya A, Rabai F, Shukla K, Doré S. Blood-Related Toxicity after Traumatic Brain Injury: Potential Targets for Neuroprotection. Mol Neurobiol 2019; 57:159-178. [PMID: 31617072 DOI: 10.1007/s12035-019-01766-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
Abstract
Emergency visits, hospitalizations, and deaths due to traumatic brain injury (TBI) have increased significantly over the past few decades. While the primary early brain trauma is highly deleterious to the brain, the secondary injury post-TBI is postulated to significantly impact mortality. The presence of blood, particularly hemoglobin, and its breakdown products and key binding proteins and receptors modulating their clearance may contribute significantly to toxicity. Heme, hemin, and iron, for example, cause membrane lipid peroxidation, generate reactive oxygen species, and sensitize cells to noxious stimuli resulting in edema, cell death, and increased morbidity and mortality. A wide range of other mechanisms such as the immune system play pivotal roles in mediating secondary injury. Effective scavenging of all of these pro-oxidant and pro-inflammatory metabolites as well as controlling maladaptive immune responses is essential for limiting toxicity and secondary injury. Hemoglobin metabolism is mediated by key molecules such as haptoglobin, heme oxygenase, hemopexin, and ferritin. Genetic variability and dysfunction affecting these pathways (e.g., haptoglobin and heme oxygenase expression) have been implicated in the difference in susceptibility of individual patients to toxicity and may be target pathways for potential therapeutic interventions in TBI. Ongoing collaborative efforts are required to decipher the complexities of blood-related toxicity in TBI with an overarching goal of providing effective treatment options to all patients with TBI.
Collapse
Affiliation(s)
- Steven A Robicsek
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA. .,Departments of Neurosurgery, Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.
| | - Ayon Bhattacharya
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA.,Department of Pharmacology, KPC Medical College, West Bengal University of Health Sciences, Kolkata, West Bengal, India
| | - Ferenc Rabai
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA
| | - Krunal Shukla
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA
| | - Sylvain Doré
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA. .,Departments of Neurology, Psychiatry, Pharmaceutics and Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
22
|
Ding M, Chen Y, Luan H, Zhang X, Zhao Z, Wu Y. Dexmedetomidine reduces inflammation in traumatic brain injury by regulating the inflammatory responses of macrophages and splenocytes. Exp Ther Med 2019; 18:2323-2331. [PMID: 31410183 PMCID: PMC6676199 DOI: 10.3892/etm.2019.7790] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 05/09/2019] [Indexed: 12/20/2022] Open
Abstract
Traumatic brain injury (TBI) affects people in all demographics, since it is associated with a variety of chronic degenerative diseases, such as Alzheimer's and Parkinson's disease. In TBI, the central nervous system elicits an immune response involving various immune cells that is necessary for healing and defending the body against pathogens, but can also cause secondary damage to the brain if the response is prolonged. In our clinical practice, it has been identified that administration of dexmedetomidine was associated with reduced production of inflammatory cytokines in patients with TBI, which led to the hypothesis that dexmedetomidine may regulate certain inflammatory responses. To test this hypothesis, the roles of dexmedetomidine in the immune system of mice were investigated. Different biological assays were used to assess the influence of dexmedetomidine on the production of inflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-8 and IL-1β. To understand how dexmedetomidine affects different types of immune cells, the influence of dexmedetomidine on splenocytes was also investigated. Finally, the effects of dexmedetomidine on macrophage activation and inflammatory functions were studied. In the present study, clinical observations and in vivo results using a mouse model of TBI revealed the regulatory functions of dexmedetomidine in TBI-associated immune response.
Collapse
Affiliation(s)
- Mengyao Ding
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Ying Chen
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Hengfei Luan
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Xiaobao Zhang
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Zhibin Zhao
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Yong Wu
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| |
Collapse
|
23
|
Cao L, Guo Y, Zhu Z. Study of the Inflammatory Mechanisms in Hyperhomocysteinemia on Large-Artery Atherosclerosis Based on Hypersensitive C-Reactive Protein-A Study from Southern China. J Stroke Cerebrovasc Dis 2019; 28:1816-1823. [PMID: 31080137 DOI: 10.1016/j.jstrokecerebrovasdis.2019.04.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/10/2019] [Accepted: 04/17/2019] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE To study the inflammatory mechanism of hyperhomocysteinemia on large-artery atherosclerosis based on hypersensitive C-reactive protein in patients. METHODS In all, 153 inpatients and 1357 physical examinees were selected. The levels of homocysteine were compared between the carotid/intracranial artery stenosis group and the nonstenosis group, between the carotid artery unstable plaque group and the nonplaque group, and between the intima-media thickness (IMT) greater than or equal to 1 group and the normal IMT group. The hypersensitive C-reactive protein levels were compared between the lacunar infarction (LI) group and the nonstroke control group and between the unstable plaque group and the nonplaque group. RESULTS Homocysteine level was significantly higher in the carotid/intracranial artery stenosis group than in the nonstenosis group, in the LI group than in the inpatient nonstroke group, and in the IMT greater than or equal to 1 group than in the normal IMT group. The hypersensitive C-reactive protein level was significantly higher in the LI group than in the nonstroke group and in the unstable plaque group than in the nonplaque group. CONCLUSIONS Hyperhomocysteinemia may aggravate the development of IMT, carotid atherosclerotic plaque instability, and carotid/intracranial artery stenosis by increasing inflammation, ultimately leading to the occurrence of LI. Hyperhomocysteinemia-induced inflammation mechanism warrants further study.
Collapse
Affiliation(s)
- Liming Cao
- Department of neurology, The First Affiliated Hospital of Jinan University, Guang zhou, China; Department of Neurology, The 3rd Affiliated Hospital of Shenzhen University, Shenzhen City, China.
| | - Yi Guo
- Department of Neurology, Shenzhen People's Hospital, Shenzhen City, China
| | - Zhishan Zhu
- Department of Neurology, The 3rd Affiliated Hospital of Shenzhen University, Shenzhen City, China
| |
Collapse
|
24
|
Abstract
Approximately 75% of patients with late-stage breast cancer will develop bone metastasis. This condition is currently considered incurable and patients' life expectancy is limited to 2-3 years following diagnosis of bone involvement. Interleukin (IL)-1B is a pro-inflammatory cytokine whose expression in primary tumours has been identified as a potential biomarker for predicting breast cancer patients at increased risk for developing bone metastasis. In this review, we discuss how IL-1B from both the tumour cells and the tumour microenvironment influence growth of primary breast tumours, dissemination into the bone metastatic niche and proliferation into overt metastases. Recent evidence indicates that targeting IL-1B signalling may provide promising new treatments that can hold tumour cells in a dormant state within bone thus preventing formation of overt bone metastases.
Collapse
Affiliation(s)
- Claudia Tulotta
- Department of Oncology and MetabolismMellanby Centre for Bone Research, University of Sheffield, Medical School, Sheffield, UK
| | - Penelope Ottewell
- Department of Oncology and MetabolismMellanby Centre for Bone Research, University of Sheffield, Medical School, Sheffield, UK
| |
Collapse
|
25
|
Letson HL, Dobson GP. Adenosine, lidocaine, and Mg2+ (ALM) resuscitation fluid protects against experimental traumatic brain injury. J Trauma Acute Care Surg 2018; 84:908-916. [PMID: 29554045 DOI: 10.1097/ta.0000000000001874] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Currently, no drug therapy prevents secondary injury progression after traumatic brain injury (TBI). Our aim was to investigate the effects of small-volume intravenous adenosine, lidocaine, and Mg (ALM) resuscitation fluid after moderate TBI in a rat fluid-percussion injury model. METHODS Anesthetized, mechanically ventilated male Sprague-Dawley rats (449 ± 5 g) were randomly assigned to one of four groups: (1) sham (craniotomy without TBI), (2) no-treatment, (3) saline-control, or (4) ALM therapy groups (all n = 16). A subdural probe was implanted in eight animals per group to measure cerebral blood flow. Fifteen minutes after moderate TBI was induced with lateral fluid percussion injury (2.57 atm), a single 3% NaCl ± ALM bolus (0.7 mL/kg) was injected intravenously, and after 60 minutes (Phase 1), 0.9% NaCl ± ALM stabilization "drip" (0.5 mL/kg per hour) was administered for 3 hours (Phase 2). RESULTS Mortality (without subdural brain probe) was 25% (saline controls) and 0% (ALM). Sixty minutes after bolus, ALM significantly increased cardiac function, cortical blood flow (CBF; approximately threefold) and blunted systemic inflammation compared to saline controls. Three hours after infusion drip, ALM improved left ventricular function, supported higher CBF, decreased proinflammatory cytokines systemically (IL-1β, tumor necrosis factor α, and regulated on activation, normal T cell expressed and secreted [RANTES]), increased anti-inflammatory cytokines in brain tissue (IL-10, IL-4), lowered brain injury markers (neuron-specific enolase, Syndecan-1, HMGB-1), reduced coagulopathy, increased platelet aggregation, and maintained baseline fibrinogen levels. Saline-controls were proinflammatory (brain, heart, lung, and blood) and hypocoagulable with neurogenic enlargement of the right side of the heart. Survival time significantly correlated with plasma neuron-specific enolase (p = 0.001) and CBF at 180 minutes (p = 0.009), and CBF correlated with brain anti-inflammatory cytokines (p = 0.001-0.034). CONCLUSION After moderate TBI, ALM resuscitation fluid increased survival and protected against early secondary injury by reducing coagulopathy, inflammation, and platelet dysfunction.
Collapse
Affiliation(s)
- Hayley L Letson
- From the Heart, Trauma and Sepsis Research Laboratory (H.LL, G.P.D.), College of Medicine and Dentistry. James Cook University, Townsville, Queensland, Australia
| | | |
Collapse
|
26
|
Barrington J, Lemarchand E, Allan SM. A brain in flame; do inflammasomes and pyroptosis influence stroke pathology? Brain Pathol 2018; 27:205-212. [PMID: 27997059 DOI: 10.1111/bpa.12476] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 12/14/2016] [Indexed: 12/21/2022] Open
Abstract
Stroke is one of the leading causes of death and disability worldwide. Inflammation plays a key role across the time course of stroke, from onset to the post-injury reparative phase days to months later. Several regulatory molecules are implicated in inflammation, but the most established inflammatory mediator of acute brain injury is the cytokine interleukin-1. Interleukin-1 is regulated by large, macromolecular complexes called inflammasomes, which play a central role in cytokine release and cell death. In this review we highlight recent advances in inflammasome research and propose key roles for inflammasome components in the progression of stroke damage.
Collapse
Affiliation(s)
- Jack Barrington
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Eloise Lemarchand
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Stuart M Allan
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, United Kingdom
| |
Collapse
|
27
|
Interleukin-1 Receptor in Seizure Susceptibility after Traumatic Injury to the Pediatric Brain. J Neurosci 2017; 37:7864-7877. [PMID: 28724747 DOI: 10.1523/jneurosci.0982-17.2017] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/29/2017] [Accepted: 07/07/2017] [Indexed: 12/19/2022] Open
Abstract
Epilepsy after pediatric traumatic brain injury (TBI) is associated with poor quality of life. This study aimed to characterize post-traumatic epilepsy in a mouse model of pediatric brain injury, and to evaluate the role of interleukin-1 (IL-1) signaling as a target for pharmacological intervention. Male mice received a controlled cortical impact or sham surgery at postnatal day 21, approximating a toddler-aged child. Mice were treated acutely with an IL-1 receptor antagonist (IL-1Ra; 100 mg/kg, s.c.) or vehicle. Spontaneous and evoked seizures were evaluated from video-EEG recordings. Behavioral assays tested for functional outcomes, postmortem analyses assessed neuropathology, and brain atrophy was detected by ex vivo magnetic resonance imaging. At 2 weeks and 3 months post-injury, TBI mice showed an elevated seizure response to the convulsant pentylenetetrazol compared with sham mice, associated with abnormal hippocampal mossy fiber sprouting. A robust increase in IL-1β and IL-1 receptor were detected after TBI. IL-1Ra treatment reduced seizure susceptibility 2 weeks after TBI compared with vehicle, and a reduction in hippocampal astrogliosis. In a chronic study, IL-1Ra-TBI mice showed improved spatial memory at 4 months post-injury. At 5 months, most TBI mice exhibited spontaneous seizures during a 7 d video-EEG recording period. At 6 months, IL-1Ra-TBI mice had fewer evoked seizures compared with vehicle controls, coinciding with greater preservation of cortical tissue. Findings demonstrate this model's utility to delineate mechanisms underlying epileptogenesis after pediatric brain injury, and provide evidence of IL-1 signaling as a mediator of post-traumatic astrogliosis and seizure susceptibility.SIGNIFICANCE STATEMENT Epilepsy is a common cause of morbidity after traumatic brain injury in early childhood. However, a limited understanding of how epilepsy develops, particularly in the immature brain, likely contributes to the lack of efficacious treatments. In this preclinical study, we first demonstrate that a mouse model of traumatic injury to the pediatric brain reproduces many neuropathological and seizure-like hallmarks characteristic of epilepsy. Second, we demonstrate that targeting the acute inflammatory response reduces cognitive impairments, the degree of neuropathology, and seizure susceptibility, after pediatric brain injury in mice. These findings provide evidence that inflammatory cytokine signaling is a key process underlying epilepsy development after an acquired brain insult, which represents a feasible therapeutic target to improve quality of life for survivors.
Collapse
|
28
|
Zhang DD, Zou MJ, Zhang YT, Fu WL, Xu T, Wang JX, Xia WR, Huang ZG, Gan XD, Zhu XM, Xu DG. A novel IL-1RA-PEP fusion protein with enhanced brain penetration ameliorates cerebral ischemia-reperfusion injury by inhibition of oxidative stress and neuroinflammation. Exp Neurol 2017; 297:1-13. [PMID: 28602833 DOI: 10.1016/j.expneurol.2017.06.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/14/2017] [Accepted: 06/07/2017] [Indexed: 12/21/2022]
Abstract
Neuroinflammation and oxidative stress are involved in cerebral ischemia-reperfusion, in which Interleukin 1 (IL-1), as an effective intervention target, is implicated. Interleukin-1 receptor antagonist (IL-1RA) is the natural inhibitor of IL-1, but blood-brain barrier (BBB) limits the brain penetration of intravenously administered IL-1RA, thereby restricting its therapeutic effect against neuroinflammation. In this study, we evaluated the potential effects of anti-inflammation and anti-oxidative stress of a novel protein IL-1RA-PEP, which fused IL-1RA with a cell penetrating peptide (CPP). Studies were carried out in transient middle cerebral artery occlusion (MCAO) in rats and oxygen glucose deprivation/reoxygenation (OGD/R) in primary cortical neurons. In MCAO rat model, IL-1RA-PEP (50mg/kg) injected i.v., penetrated BBB effectively, and alleviated brain infarction, cerebral edema, neurological deficit score and motor performance as well as inhibited the inflammatory cytokines expression. Furthermore, our results firstly showed that IL-1RA-PEP also regulated the oxidases expression, decreased the levels of NO, MDA and ROS. In addition, the inhibitory effects of IL-1RA-PEP on oxidative stress and inflammation were confirmed in rat cortical neurons induced by OGD/R, it reduced ROS, IL-6 and TNF-α. Further study showed that the effects of IL-1RA-PEP were closely associated with the NF-κB and p38 pathways which were proved respectively by their inhibitors JSH-23 and SB203580. Our results indicated that IL-1RA-PEP could effectively penetrate the brain of MCAO rats, alleviated the cerebral ischemia reperfusion injury by inhibiting neuroinflammation and oxidative stress, showing a great clinical potential for stroke.
Collapse
Affiliation(s)
- Dong-Dong Zhang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China; Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China
| | - Min-Ji Zou
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Ya-Tao Zhang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Wen-Liang Fu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Tao Xu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Jia-Xi Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Wen-Rong Xia
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Zhi-Guang Huang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Xiang-Dong Gan
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Xiao-Ming Zhu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Dong-Gang Xu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China; Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China.
| |
Collapse
|
29
|
Zhao Q, Che X, Zhang H, Fan P, Tan G, Liu L, Jiang D, Zhao J, Xiang X, Liang Y, Sun X, He Z. Thioredoxin-interacting protein links endoplasmic reticulum stress to inflammatory brain injury and apoptosis after subarachnoid haemorrhage. J Neuroinflammation 2017; 14:104. [PMID: 28490373 PMCID: PMC5426069 DOI: 10.1186/s12974-017-0878-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 05/01/2017] [Indexed: 02/05/2023] Open
Abstract
Background Early brain injury (EBI) is considered a major contributor to the high morbidity and mortality associated with subarachnoid haemorrhage (SAH). Both of sterile inflammation and apoptosis are considered the important causes of EBI. Recently, it was confirmed that thioredoxin-interacting protein (TXNIP) not only participates in inflammatory amplification but also stimulates the apoptosis signalling cascade pathway. However, whether the effects of TXNIP influence the pathogenesis of SAH remains unclear. Here, we hypothesize that TXNIP activity induced by endoplasmic reticulum stress (ER stress) may contribute to the pathogenesis of EBI through pro-inflammatory and pro-apoptotic mechanisms. Methods A total of 299 male Sprague–Dawley rats were used to create SAH models. Resveratrol (RES, 60 mg/kg) and two TXNIP small interfering RNA (siRNA) were used to inhibit TXNIP expression. The specific inhibitors of ER stress sensors were used to disrupt the link between TXNIP and ER stress. SAH grade, neurological deficits, brain water content and blood–brain barrier (BBB) permeability were evaluated simultaneously as prognostic indicators. Fluorescent double-labelling was employed to detect the location of TXNIP in cerebral cells. Western blot and TUNEL were performed to study the mechanisms of TXNIP and EBI. Results We found that TXNIP expression significantly increased after SAH, peaking at 48 h (0.48 ± 0.04, up to 3.2-fold) and decreasing at 72 h after surgery. This process was accompanied by the generation of inflammation-associated factors. TXNIP was expressed in the cytoplasm of neurons and was widely co-localized with TUNEL-positive cells in both the hippocampus and the cortex of SAH rats. We discovered for the first time that TXNIP was co-localized in neural immunocytes (microglia and astrocytes). After administration of RES, TXNIP siRNA and ER stress inhibitors, TXNIP expression was significantly reduced and the crosstalk between TXNIP and ER stress was disrupted; this was accompanied by a reduction in inflammatory and apoptotic factors, as well as attenuation of the prognostic indices. Conclusions These results may represent the critical evidence to support the pro-inflammatory and pro-apoptotic effects of TXNIP after SAH. Our data suggest that TXNIP participates in EBI after SAH by mediating inflammation and apoptosis; these pathways may represent a potential therapeutic strategy for SAH treatment. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0878-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qing Zhao
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, 400016, Chongqing, China
| | - Xudong Che
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, 400016, Chongqing, China
| | - Hongxia Zhang
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, 400016, Chongqing, China
| | - Pianpian Fan
- Department of Endocrinology, West China Hospital of Sichuan University, 37 Guo Xue Xiang, 610041, Chengdu, Sichuan, China
| | - Guanping Tan
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, 400016, Chongqing, China
| | - Liu Liu
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, 400016, Chongqing, China
| | - Dengzhi Jiang
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, 400016, Chongqing, China
| | - Jun Zhao
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, 400016, Chongqing, China
| | - Xiang Xiang
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, 400016, Chongqing, China
| | - Yidan Liang
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, 400016, Chongqing, China
| | - Xiaochuan Sun
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, 400016, Chongqing, China
| | - Zhaohui He
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, 400016, Chongqing, China.
| |
Collapse
|
30
|
Gougeon ML. Alarmins and central nervous system inflammation in HIV-associated neurological disorders. J Intern Med 2017; 281:433-447. [PMID: 27862491 DOI: 10.1111/joim.12570] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In the era of highly active antiretroviral therapy (HAART), HIV-1-associated neurocognitive disorders (HAND) persist in infected individuals with adequate immunological and virological status. Risk factors for cognitive impairment include hepatitis C virus co-infection, host genetic factors predisposing to HAND, the early establishment of the virus in the CNS and its persistence under HAART; thus, the CNS is an important reservoir for HIV. Microglial cells are permissive to HIV-1, and NLRP3 inflammasome-associated genes were found expressed in brains of HIV-1-infected persons, contributing to brain disease. Inflammasomes can be triggered by alarmins or danger-associated molecular patterns (DAMPs), which directly stimulate the production of proinflammatory mediators by glial cells, contribute to blood-brain barrier injury through induction of release of various proteases and allow the passage of infected macrophages, and trigger IL-1β release from primed cells. Amongst alarmins involved in HIV-1-induced neuropathogenesis, IL-33 and high-mobility group box 1 (HMGB1) are of particular interest. Neurocognitive alterations were recently associated with dysregulation of the IL-33/ST2 axis in the CNS, leading to the induction of neuronal apoptosis, decrease in synaptic function and neuroinflammation. Specific biomarkers, including HMGB1 and anti-HMGB1 antibodies, have been identified in cerebrospinal fluid from patients with HAND, correlated with immune activation and identifying a very early stage of neurocognitive impairment that precedes changes in metabolites detected by magnetic resonance spectroscopy. Moreover, HMGB1 plays a crucial role in HIV-1 persistence in dendritic cells and in the constitution of viral reservoirs. In this review, the mechanisms whereby alarmins contribute to HIV-1-induced CNS inflammation and neuropathogenesis will be discussed.
Collapse
Affiliation(s)
- M-L Gougeon
- Institut Pasteur, Antiviral Immunity, Biotherapy and Vaccine Unit, Infection and Epidemiology Department, Paris, France
| |
Collapse
|
31
|
Dickens AM, Tovar-Y-Romo LB, Yoo SW, Trout AL, Bae M, Kanmogne M, Megra B, Williams DW, Witwer KW, Gacias M, Tabatadze N, Cole RN, Casaccia P, Berman JW, Anthony DC, Haughey NJ. Astrocyte-shed extracellular vesicles regulate the peripheral leukocyte response to inflammatory brain lesions. Sci Signal 2017; 10:10/473/eaai7696. [PMID: 28377412 PMCID: PMC5590230 DOI: 10.1126/scisignal.aai7696] [Citation(s) in RCA: 199] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Brain injury induces a peripheral acute cytokine response that directs the transmigration of leukocytes into the brain. Because this brain-to-peripheral immune communication affects patient recovery, understanding its regulation is important. Using a mouse model of inflammatory brain injury, we set out to find a soluble mediator for this phenomenon. We found that extracellular vesicles (EVs) shed from astrocytes in response to intracerebral injection of interleukin-1β (IL-1β) rapidly entered into peripheral circulation and promoted the transmigration of leukocytes through modulation of the peripheral acute cytokine response. Bioinformatic analysis of the protein and microRNA cargo of EVs identified peroxisome proliferator-activated receptor α (PPARα) as a primary molecular target of astrocyte-shed EVs. We confirmed in mice that astrocytic EVs promoted the transmigration of leukocytes into the brain by inhibiting PPARα, resulting in the increase of nuclear factor κB (NF-κB) activity that triggered the production of cytokines in liver. These findings expand our understanding of the mechanisms regulating communication between the brain and peripheral immune system and identify astrocytic EVs as a molecular regulator of the immunological response to inflammatory brain damage.
Collapse
Affiliation(s)
- Alex M Dickens
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Luis B Tovar-Y-Romo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Seung-Wan Yoo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Amanda L Trout
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mihyun Bae
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Marlene Kanmogne
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Bezawit Megra
- Departments of Pathology, Microbiology, and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Dionna W Williams
- Departments of Pathology, Microbiology, and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kennith W Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mar Gacias
- Department of Neuroscience, Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nino Tabatadze
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Robert N Cole
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Patrizia Casaccia
- Department of Neuroscience, Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joan W Berman
- Departments of Pathology, Microbiology, and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Daniel C Anthony
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Norman J Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA. .,Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
32
|
Conos SA, Chen KW, De Nardo D, Hara H, Whitehead L, Núñez G, Masters SL, Murphy JM, Schroder K, Vaux DL, Lawlor KE, Lindqvist LM, Vince JE. Active MLKL triggers the NLRP3 inflammasome in a cell-intrinsic manner. Proc Natl Acad Sci U S A 2017; 114:E961-E969. [PMID: 28096356 PMCID: PMC5307433 DOI: 10.1073/pnas.1613305114] [Citation(s) in RCA: 320] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Necroptosis is a physiological cell suicide mechanism initiated by receptor-interacting protein kinase-3 (RIPK3) phosphorylation of mixed-lineage kinase domain-like protein (MLKL), which results in disruption of the plasma membrane. Necroptotic cell lysis, and resultant release of proinflammatory mediators, is thought to cause inflammation in necroptotic disease models. However, we previously showed that MLKL signaling can also promote inflammation by activating the nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome to recruit the adaptor protein apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC) and trigger caspase-1 processing of the proinflammatory cytokine IL-1β. Here, we provide evidence that MLKL-induced activation of NLRP3 requires (i) the death effector four-helical bundle of MLKL, (ii) oligomerization and association of MLKL with cellular membranes, and (iii) a reduction in intracellular potassium concentration. Although genetic or pharmacological targeting of NLRP3 or caspase-1 prevented MLKL-induced IL-1β secretion, they did not prevent necroptotic cell death. Gasdermin D (GSDMD), the pore-forming caspase-1 substrate required for efficient NLRP3-triggered pyroptosis and IL-1β release, was not essential for MLKL-dependent death or IL-1β secretion. Imaging of MLKL-dependent ASC speck formation demonstrated that necroptotic stimuli activate NLRP3 cell-intrinsically, indicating that MLKL-induced NLRP3 inflammasome formation and IL-1β cleavage occur before cell lysis. Furthermore, we show that necroptotic activation of NLRP3, but not necroptotic cell death alone, is necessary for the activation of NF-κB in healthy bystander cells. Collectively, these results demonstrate the potential importance of NLRP3 inflammasome activity as a driving force for inflammation in MLKL-dependent diseases.
Collapse
Affiliation(s)
- Stephanie A Conos
- Cell Signalling and Cell Death Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Kaiwen W Chen
- Institute for Molecular Bioscience and Centre for Inflammation and Disease Research, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Dominic De Nardo
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Hideki Hara
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Lachlan Whitehead
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
- Systems Biology & Personalised Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Gabriel Núñez
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Seth L Masters
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - James M Murphy
- Cell Signalling and Cell Death Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Kate Schroder
- Institute for Molecular Bioscience and Centre for Inflammation and Disease Research, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - David L Vaux
- Cell Signalling and Cell Death Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Kate E Lawlor
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia;
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Lisa M Lindqvist
- Cell Signalling and Cell Death Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia;
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - James E Vince
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia;
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| |
Collapse
|
33
|
Shukla V, Shakya AK, Perez-Pinzon MA, Dave KR. Cerebral ischemic damage in diabetes: an inflammatory perspective. J Neuroinflammation 2017; 14:21. [PMID: 28115020 PMCID: PMC5260103 DOI: 10.1186/s12974-016-0774-5] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 12/07/2016] [Indexed: 12/16/2022] Open
Abstract
Stroke is one of the leading causes of death worldwide. A strong inflammatory response characterized by activation and release of cytokines, chemokines, adhesion molecules, and proteolytic enzymes contributes to brain damage following stroke. Stroke outcomes are worse among diabetics, resulting in increased mortality and disabilities. Diabetes involves chronic inflammation manifested by reactive oxygen species generation, expression of proinflammatory cytokines, and activation/expression of other inflammatory mediators. It appears that increased proinflammatory processes due to diabetes are further accelerated after cerebral ischemia, leading to increased ischemic damage. Hypoglycemia is an intrinsic side effect owing to glucose-lowering therapy in diabetics, and is known to induce proinflammatory changes as well as exacerbate cerebral damage in experimental stroke. Here, we present a review of available literature on the contribution of neuroinflammation to increased cerebral ischemic damage in diabetics. We also describe the role of hypoglycemia in neuroinflammation and cerebral ischemic damage in diabetics. Understanding the role of neuroinflammatory mechanisms in worsening stroke outcome in diabetics may help limit ischemic brain injury and improve clinical outcomes.
Collapse
Affiliation(s)
- Vibha Shukla
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, Miami, FL, 33136, USA.,Department of Neurology (D4-5), University of Miami Miller School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA
| | - Akhalesh Kumar Shakya
- Present address: Department of Microbiology and Immunology, and Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130, USA
| | - Miguel A Perez-Pinzon
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, Miami, FL, 33136, USA.,Department of Neurology (D4-5), University of Miami Miller School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Neuroscience Program, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Kunjan R Dave
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, Miami, FL, 33136, USA. .,Department of Neurology (D4-5), University of Miami Miller School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA. .,Neuroscience Program, University of Miami School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
34
|
Amiri S, Haj-Mirzaian A, Momeny M, Amini-Khoei H, Rahimi-Balaei M, Poursaman S, Rastegar M, Nikoui V, Mokhtari T, Ghazi-Khansari M, Hosseini MJ. Streptozotocin induced oxidative stress, innate immune system responses and behavioral abnormalities in male mice. Neuroscience 2017; 340:373-383. [DOI: 10.1016/j.neuroscience.2016.11.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 11/03/2016] [Accepted: 11/04/2016] [Indexed: 01/08/2023]
|
35
|
Brickler T, Gresham K, Meza A, Coutermarsh-Ott S, Williams TM, Rothschild DE, Allen IC, Theus MH. Nonessential Role for the NLRP1 Inflammasome Complex in a Murine Model of Traumatic Brain Injury. Mediators Inflamm 2016; 2016:6373506. [PMID: 27199506 PMCID: PMC4854993 DOI: 10.1155/2016/6373506] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/08/2016] [Accepted: 03/24/2016] [Indexed: 01/01/2023] Open
Abstract
Traumatic brain injury (TBI) elicits the immediate production of proinflammatory cytokines which participate in regulating the immune response. While the mechanisms of adaptive immunity in secondary injury are well characterized, the role of the innate response is unclear. Recently, the NLR inflammasome has been shown to become activated following TBI, causing processing and release of interleukin-1β (IL-1β). The inflammasome is a multiprotein complex consisting of nucleotide-binding domain and leucine-rich repeat containing proteins (NLR), caspase-1, and apoptosis-associated speck-like protein (ASC). ASC is upregulated after TBI and is critical in coupling the proteins during complex formation resulting in IL-1β cleavage. To directly test whether inflammasome activation contributes to acute TBI-induced damage, we assessed IL-1β, IL-18, and IL-6 expression, contusion volume, hippocampal cell death, and motor behavior recovery in Nlrp1(-/-), Asc(-/-), and wild type mice after moderate controlled cortical impact (CCI) injury. Although IL-1β expression is significantly attenuated in the cortex of Nlrp1(-/-) and Asc(-/-) mice following CCI injury, no difference in motor recovery, cell death, or contusion volume is observed compared to wild type. These findings indicate that inflammasome activation does not significantly contribute to acute neural injury in the murine model of moderate CCI injury.
Collapse
Affiliation(s)
- Thomas Brickler
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Duck Pond Drive, Blacksburg, VA 24061, USA
| | - Kisha Gresham
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Duck Pond Drive, Blacksburg, VA 24061, USA
| | - Armand Meza
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Duck Pond Drive, Blacksburg, VA 24061, USA
| | - Sheryl Coutermarsh-Ott
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Duck Pond Drive, Blacksburg, VA 24061, USA
| | - Tere M. Williams
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Duck Pond Drive, Blacksburg, VA 24061, USA
| | - Daniel E. Rothschild
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Duck Pond Drive, Blacksburg, VA 24061, USA
| | - Irving C. Allen
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Duck Pond Drive, Blacksburg, VA 24061, USA
| | - Michelle H. Theus
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Duck Pond Drive, Blacksburg, VA 24061, USA
| |
Collapse
|
36
|
Fluoxetine protects against IL-1β-induced neuronal apoptosis via downregulation of p53. Neuropharmacology 2016; 107:68-78. [PMID: 26976669 DOI: 10.1016/j.neuropharm.2016.03.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/02/2016] [Accepted: 03/10/2016] [Indexed: 12/23/2022]
Abstract
Fluoxetine, a selective serotonin reuptake inhibitor, exerts neuroprotective effects in a variety of neurological diseases including stroke, but the underlying mechanism remains obscure. In the present study, we addressed the molecular events in fluoxetine against ischemia/reperfusion-induced acute neuronal injury and inflammation-induced neuronal apoptosis. We showed that treatment of fluoxetine (40 mg/kg, i.p.) with twice injections at 1 h and 12 h after transient middle cerebral artery occlusion (tMCAO) respectively alleviated neurological deficits and neuronal apoptosis in a mouse ischemic stroke model, accompanied by inhibiting interleukin-1β (IL-1β), Bax and p53 expression and upregulating anti-apoptotic protein Bcl-2 level. We next mimicked neuroinflammation in ischemic stroke with IL-1β in primary cultured cortical neurons and found that pretreatment with fluoxetine (1 μM) prevented IL-1β-induced neuronal apoptosis and upregulation of p53 expression. Furthermore, we demonstrated that p53 overexpression in N2a cell line abolished the anti-apoptotic effect of fluoxetine, indicating that p53 downregulation is required for the protective role of fluoxetine in IL-1β-induced neuronal apoptosis. Fluoxetine downregulating p53 expression could be mimicked by SB203580, a specific inhibitor of p38, but blocked by anisomycin, a p38 activator. Collectively, our findings have revealed that fluoxetine protects against IL-1β-induced neuronal apoptosis via p38-p53 dependent pathway, which give us an insight into the potential of fluoxetine in terms of opening up novel therapeutic avenues for neurological diseases including stroke.
Collapse
|
37
|
Xing Z, Xia Z, Peng W, Li J, Zhang C, Fu C, Tang T, Luo J, Zou Y, Fan R, Liu W, Xiong X, Huang W, Sheng C, Gan P, Wang Y. Xuefu Zhuyu decoction, a traditional Chinese medicine, provides neuroprotection in a rat model of traumatic brain injury via an anti-inflammatory pathway. Sci Rep 2016; 6:20040. [PMID: 26818584 PMCID: PMC4730240 DOI: 10.1038/srep20040] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/23/2015] [Indexed: 12/28/2022] Open
Abstract
Neuroinflammation is central to the pathology of traumatic brain injury (TBI). Xuefu Zhuyu decoction (XFZY) is an effective traditional Chinese medicine to treat TBI. To elucidate its potential molecular mechanism, this study aimed to demonstrate that XFZY functions as an anti-inflammatory agent by inhibiting the PI3K-AKT-mTOR pathway. Sprague-Dawley rats were exposed to controlled cortical impact to produce a neuroinflammatory response. The treatment groups received XFZY (9 g/kg and 18 g/kg), Vehicle group and Sham group were gavaged with equal volumes of saline. The modified neurologic severity score (mNSS) and the Morris water maze test were used to assess neurological deficits. Arachidonic acid (AA) levels in brain tissue were measured using tandem gas chromatography-mass spectrometry. TNF-α and IL-1β levels in injured ipsilateral brain tissue were detected by ELISA. AKT and mTOR expression were measured by western blot analysis. The results indicated that XFZY significantly enhanced spatial memory acquisition. XFZY (especially at a dose of 9 g/kg) markedly reduced the mNSS and levels of AA, TNF-α and IL-1β. Significant downregulation of AKT/mTOR/p70S6K proteins in brain tissues was observed after the administration of XFZY (especially at a dose of 9 g/kg). XFZY may be a promising therapeutic strategy for reducing inflammation in TBI.
Collapse
Affiliation(s)
- Zhihua Xing
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Zian Xia
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Weijun Peng
- Department of traditional Chinese medicine, 2nd Xiangya Hospital, Central South University, 410011 Changsha, China
| | - Jun Li
- Thyroid Tumour Internal Medicine Department, Cancer Hospital affiliated to Xiangya School of Medicine, Central South University, 410013 Changsha, China
| | - Chunhu Zhang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Chunyan Fu
- Department of Pharmacy, Shaoyang Medical College Level Specialty School, 422000 Shaoyang, China
| | - Tao Tang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Jiekun Luo
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Yong Zou
- Department of Gerontology and Respiratory Diseases, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Rong Fan
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Weiping Liu
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Xingui Xiong
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Wei Huang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Chenxia Sheng
- Department of traditional Chinese medicine, 2nd Xiangya Hospital, Central South University, 410011 Changsha, China
| | - Pingping Gan
- Department of Oncology, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Yang Wang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| |
Collapse
|
38
|
Therapies negating neuroinflammation after brain trauma. Brain Res 2015; 1640:36-56. [PMID: 26740405 DOI: 10.1016/j.brainres.2015.12.024] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 12/07/2015] [Accepted: 12/14/2015] [Indexed: 12/11/2022]
Abstract
Traumatic brain injury (TBI) elicits a complex secondary injury response, with neuroinflammation as a crucial central component. Long thought to be solely a deleterious factor, the neuroinflammatory response has recently been shown to be far more intricate, with both beneficial and detrimental consequences depending on the timing, magnitude and specific immune composition of the response post-injury. Despite extensive preclinical and clinical research into mechanisms of secondary injury after TBI, no effective neuroprotective therapy has been identified, with potential candidates repeatedly proving disappointing in the clinic. The neuroinflammatory response offers a promising avenue for therapeutic targeting, aiming to quell the deleterious consequences without influencing its function in providing a neurotrophic environment supportive of repair. The present review firstly describes the findings of recent clinical trials that aimed to modulate inflammation as a means of neuroprotection. Secondly, we discuss promising multifunctional and single-target anti-inflammatory candidates either currently in trial, or with ample experimental evidence supporting clinical application. This article is part of a Special Issue entitled SI:Brain injury and recovery.
Collapse
|
39
|
Ydens E, Demon D, Lornet G, De Winter V, Timmerman V, Lamkanfi M, Janssens S. Nlrp6 promotes recovery after peripheral nerve injury independently of inflammasomes. J Neuroinflammation 2015; 12:143. [PMID: 26253422 PMCID: PMC4528710 DOI: 10.1186/s12974-015-0367-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 07/24/2015] [Indexed: 11/19/2022] Open
Abstract
Background NOD-like receptors (Nlrs) are key regulators of immune responses during infection and autoimmunity. A subset of Nlrs assembles inflammasomes, molecular platforms that are activated in response to endogenous danger and microbial ligands and that control release of interleukin (IL)-1β and IL-18. However, their role in response to injury in the nervous system is less understood. Methods In this study, we investigated the expression profile of major inflammasome components in the peripheral nervous system (PNS) and explored the physiological role of different Nlrs upon acute nerve injury in mice. Results While in basal conditions, predominantly members of NOD-like receptor B (Nlrb) subfamily (NLR family, apoptosis inhibitory proteins (NAIPs)) and Nlrc subfamily (ICE-protease activating factor (IPAF)/NOD) are detected in the sciatic nerve, injury causes a shift towards expression of the Nlrp family. Sterile nerve injury also leads to an increase in expression of the Nlrb subfamily, while bacteria trigger expression of the Nlrc subfamily. Interestingly, loss of Nlrp6 led to strongly impaired nerve function upon nerve crush. Loss of the inflammasome adaptor apoptosis-associated speck-like protein containing a CARD (ASC) and effector caspase-1 and caspase-11 did not affect sciatic nerve function, suggesting that Nlrp6 contributed to recovery after peripheral nerve injury independently of inflammasomes. In line with this, we did not detect release of mature IL-1β upon acute nerve injury despite potent induction of pro-IL-1β and inflammasome components Nlrp3 and Nlrp1. However, Nlrp6 deficiency was associated with increased pro-inflammatory extracellular regulated MAP kinase (ERK) signaling, suggesting that hyperinflammation in the absence of Nlrp6 exacerbated peripheral nerve injury. Conclusions Together, our observations suggest that Nlrp6 contributes to recovery from peripheral nerve injury by dampening inflammatory responses independently of IL-1β and inflammasomes.
Collapse
Affiliation(s)
- Elke Ydens
- Peripheral Neuropathy Group, Department of Molecular Genetics, VIB and University of Antwerp, Universiteitsplein 1, B-2610, Wilrijk, Antwerpen, Belgium. .,Neurogenetics Laboratory, Institute Born-Bunge and University of Antwerp, Universiteitsplein 1, B-2610, Antwerpen, Belgium.
| | - Dieter Demon
- Department of Medical Protein Research, VIB, Gent, Belgium. .,Department of Biochemistry, Ghent University, Gent, Belgium.
| | - Guillaume Lornet
- Unit Immunoregulation and Mucosal Immunology, GROUP-ID Consortium, VIB Inflammation Research Centre, Technologiepark 927, B-9052, Gent, Belgium. .,Department of Internal Medicine, Ghent University, Gent, Belgium.
| | - Vicky De Winter
- Peripheral Neuropathy Group, Department of Molecular Genetics, VIB and University of Antwerp, Universiteitsplein 1, B-2610, Wilrijk, Antwerpen, Belgium. .,Neurogenetics Laboratory, Institute Born-Bunge and University of Antwerp, Universiteitsplein 1, B-2610, Antwerpen, Belgium.
| | - Vincent Timmerman
- Peripheral Neuropathy Group, Department of Molecular Genetics, VIB and University of Antwerp, Universiteitsplein 1, B-2610, Wilrijk, Antwerpen, Belgium. .,Neurogenetics Laboratory, Institute Born-Bunge and University of Antwerp, Universiteitsplein 1, B-2610, Antwerpen, Belgium.
| | - Mohamed Lamkanfi
- Department of Medical Protein Research, VIB, Gent, Belgium. .,Department of Biochemistry, Ghent University, Gent, Belgium.
| | - Sophie Janssens
- Unit Immunoregulation and Mucosal Immunology, GROUP-ID Consortium, VIB Inflammation Research Centre, Technologiepark 927, B-9052, Gent, Belgium. .,Department of Internal Medicine, Ghent University, Gent, Belgium.
| |
Collapse
|
40
|
Xia YY, Song SW, Min Y, Zhong Y, Sheng YC, Li RP, Liu QH. The effects of anakinra on focal cerebral ischemic injury in rats. CNS Neurosci Ther 2015; 20:879-81. [PMID: 25130727 DOI: 10.1111/cns.12310] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 07/15/2014] [Accepted: 07/16/2014] [Indexed: 11/28/2022] Open
|
41
|
AIM2 and NLRC4 inflammasomes contribute with ASC to acute brain injury independently of NLRP3. Proc Natl Acad Sci U S A 2015; 112:4050-5. [PMID: 25775556 DOI: 10.1073/pnas.1419090112] [Citation(s) in RCA: 201] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Inflammation that contributes to acute cerebrovascular disease is driven by the proinflammatory cytokine interleukin-1 and is known to exacerbate resulting injury. The activity of interleukin-1 is regulated by multimolecular protein complexes called inflammasomes. There are multiple potential inflammasomes activated in diverse diseases, yet the nature of the inflammasomes involved in brain injury is currently unknown. Here, using a rodent model of stroke, we show that the NLRC4 (NLR family, CARD domain containing 4) and AIM2 (absent in melanoma 2) inflammasomes contribute to brain injury. We also show that acute ischemic brain injury is regulated by mechanisms that require ASC (apoptosis-associated speck-like protein containing a CARD), a common adaptor protein for several inflammasomes, and that the NLRP3 (NLR family, pyrin domain containing 3) inflammasome is not involved in this process. These discoveries identify the NLRC4 and AIM2 inflammasomes as potential therapeutic targets for stroke and provide new insights into how the inflammatory response is regulated after an acute injury to the brain.
Collapse
|
42
|
Ainscough JS, Frank Gerberick G, Zahedi-Nejad M, Lopez-Castejon G, Brough D, Kimber I, Dearman RJ. Dendritic cell IL-1α and IL-1β are polyubiquitinated and degraded by the proteasome. J Biol Chem 2014; 289:35582-92. [PMID: 25371210 PMCID: PMC4271241 DOI: 10.1074/jbc.m114.595686] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
IL-1α and β are key players in the innate immune system. The secretion of these cytokines by dendritic cells (DC) is integral to the development of proinflammatory responses. These cytokines are not secreted via the classical secretory pathway. Instead, 2 independent processes are required; an initial signal to induce up-regulation of the precursor pro-IL-1α and -β, and a second signal to drive cleavage and consequent secretion. Pro-IL-1α and -β are both cytosolic and thus, are potentially subject to post-translational modifications. These modifications may, in turn, have a functional outcome in the context of IL-1α and -β secretion and hence inflammation. We report here that IL-1α and -β were degraded intracellularly in murine bone marrow-derived DC and that this degradation was dependent on active cellular processes. In addition, we demonstrate that degradation was ablated when the proteasome was inhibited, whereas autophagy did not appear to play a major role. Furthermore, inhibition of the proteasome led to an accumulation of polyubiquitinated IL-1α and -β, indicating that IL-1α and -β were polyubiquitinated prior to proteasomal degradation. Finally, our investigations suggest that polyubiquitination and proteasomal degradation are not continuous processes but instead are up-regulated following DC activation. Overall, these data highlight that IL-1α and -β polyubiquitination and proteasomal degradation are central mechanisms in the regulation of intracellular IL-1 levels in DC.
Collapse
Affiliation(s)
- Joseph S Ainscough
- From the Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom and
| | | | - Maryam Zahedi-Nejad
- From the Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom and
| | - Gloria Lopez-Castejon
- From the Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom and
| | - David Brough
- From the Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom and
| | - Ian Kimber
- From the Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom and
| | - Rebecca J Dearman
- From the Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom and
| |
Collapse
|
43
|
Ogryzko NV, Renshaw SA, Wilson HL. The IL-1 family in fish: swimming through the muddy waters of inflammasome evolution. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 46:53-62. [PMID: 24690566 DOI: 10.1016/j.dci.2014.03.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/13/2014] [Accepted: 03/14/2014] [Indexed: 06/03/2023]
Abstract
Inflammatory diseases are a significant burden on global healthcare systems, and tackling these diseases is a major focus of modern medicine. Key to many inflammatory diseases is the cytokine, Interleukin-1 (IL-1). Due to its apical role in initiating the inflammatory response, dysregulated IL-1 signalling results in a number of pathologies. Treatment of inflammatory diseases with anti-IL-1 therapies has offered many therapeutic benefits, however current therapies are protein based, with all the accompanying limitations. The non-conventional pathways involved in IL-1 signalling provide a number of potential therapeutic targets for clinical intervention and this has led to the exploitation of a number of model organisms for the study of IL-1 biology. Murine models have long been used to study IL-1 processing and release, but do not allow direct visualisation in vivo. Recently, fish models have emerged as genetically tractable and optically transparent inflammatory disease models. These models have raised questions on the evolutionary origins of the IL-1 family and the conservation in its processing and activation. Here we review the current understanding of IL-1 evolution in fish and discuss the study of IL-1 processing in these models.
Collapse
Affiliation(s)
- Nikolay V Ogryzko
- Medical Research Council Centre for Developmental and Biomedical Genetics, Firth Court, University of Sheffield, Sheffield, United Kingdom; Department of Cardiovascular Science, University of Sheffield, Sheffield, United Kingdom.
| | - Stephen A Renshaw
- Medical Research Council Centre for Developmental and Biomedical Genetics, Firth Court, University of Sheffield, Sheffield, United Kingdom; Department of Infection and Immunity and MRC Centre for Developmental and Biomedical Genetics, Firth Court, University of Sheffield, Sheffield, United Kingdom
| | - Heather L Wilson
- Department of Cardiovascular Science, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
44
|
England H, Summersgill HR, Edye ME, Rothwell NJ, Brough D. Release of interleukin-1α or interleukin-1β depends on mechanism of cell death. J Biol Chem 2014; 289:15942-50. [PMID: 24790078 PMCID: PMC4047367 DOI: 10.1074/jbc.m114.557561] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The cytokine interleukin-1 (IL-1) has two main pro-inflammatory forms, IL-1α and IL-1β, which are central to host responses to infection and to damaging sterile inflammation. Processing of IL-1 precursor proteins to active cytokines commonly occurs through activation of proteases, notably caspases and calpains. These proteases are instrumental in cell death, and inflammation and cell death are closely associated, hence we sought to determine the impact of cell death pathways on IL-1 processing and release. We discovered that apoptotic regulation of caspase-8 specifically induced the processing and release of IL-1β. Conversely, necroptosis caused the processing and release of IL-1α, and this was independent of IL-1β processing and release. These data suggest that the mechanism through which an IL-1-expressing cell dies dictates the nature of the inflammatory mechanism that follows. These insights may allow modification of inflammation through the selective targeting of cell death mechanisms during disease.
Collapse
Affiliation(s)
- Hazel England
- From the Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Holly R Summersgill
- From the Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Michelle E Edye
- From the Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Nancy J Rothwell
- From the Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester M13 9PT, United Kingdom
| | - David Brough
- From the Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester M13 9PT, United Kingdom
| |
Collapse
|
45
|
Wang CP, Zhang LZ, Li GC, Shi YW, Li JL, Zhang XC, Wang ZW, Ding F, Liang XM. Mulberroside a protects against ischemic impairment in primary culture of rat cortical neurons after oxygen-glucose deprivation followed by reperfusion. J Neurosci Res 2014; 92:944-54. [DOI: 10.1002/jnr.23374] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 12/27/2013] [Accepted: 01/27/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Cai-Ping Wang
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
| | - Lu-Zhong Zhang
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
| | - Gui-Cai Li
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
| | - Yun-wei Shi
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
| | - Jian-Long Li
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
| | - Xiao-Chuan Zhang
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
| | - Zhi-Wei Wang
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
- Department of Pharmacology; University of California; Irvine California
| | - Fei Ding
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
| | - Xin-Miao Liang
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
- Dalian Institute of Chemical Physics; Chinese Academy of Sciences; Dalian China
| |
Collapse
|
46
|
Singh N, Hopkins SJ, Hulme S, Galea JP, Hoadley M, Vail A, Hutchinson PJ, Grainger S, Rothwell NJ, King AT, Tyrrell PJ. The effect of intravenous interleukin-1 receptor antagonist on inflammatory mediators in cerebrospinal fluid after subarachnoid haemorrhage: a phase II randomised controlled trial. J Neuroinflammation 2014; 11:1. [PMID: 24383930 PMCID: PMC3892121 DOI: 10.1186/1742-2094-11-1] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 12/17/2013] [Indexed: 12/12/2022] Open
Abstract
Background Interleukin-1 (IL-1) is a key mediator of ischaemic brain injury induced by stroke and subarachnoid haemorrhage (SAH). IL-1 receptor antagonist (IL-1Ra) limits brain injury in experimental stroke and reduces plasma inflammatory mediators associated with poor outcome in ischaemic stroke patients. Intravenous (IV) IL-1Ra crosses the blood–brain barrier (BBB) in patients with SAH, to achieve cerebrospinal fluid (CSF) concentrations that are neuroprotective in rats. Methods A small phase II, double-blind, randomised controlled study was carried out across two UK neurosurgical centres with the aim of recruiting 32 patients. Adult patients with aneurysmal SAH, requiring external ventricular drainage (EVD) within 72 hours of ictus, were eligible. Patients were randomised to receive IL-1Ra (500 mg bolus, then a 10 mg/kg/hr infusion for 24 hours) or placebo. Serial samples of CSF and plasma were taken and analysed for inflammatory mediators, with change in CSF IL-6 between 6 and 24 hours as the primary outcome measure. Results Six patients received IL-1Ra and seven received placebo. Concentrations of IL-6 in CSF and plasma were reduced by one standard deviation in the IL-1Ra group compared to the placebo group, between 6 and 24 hours, as predicted by the power calculation. This did not reach statistical significance (P = 0.08 and P = 0.06, respectively), since recruitment did not reach the target figure of 32. No adverse or serious adverse events reported were attributable to IL-1Ra. Conclusions IL-1Ra appears safe in SAH patients. The concentration of IL-6 was lowered to the degree expected, in both CSF and plasma for patients treated with IL-1Ra.
Collapse
Affiliation(s)
| | - Stephen J Hopkins
- The University of Manchester Stroke and Vascular Centre, Manchester Academic Health Sciences Centre, Clinical Sciences Building, Salford Royal NHS Foundation Trust, Stott Lane, Salford M6 8HD, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Prins M, Eriksson C, Wierinckx A, Bol JGJM, Binnekade R, Tilders FJH, Van Dam AM. Interleukin-1β and interleukin-1 receptor antagonist appear in grey matter additionally to white matter lesions during experimental multiple sclerosis. PLoS One 2013; 8:e83835. [PMID: 24376764 PMCID: PMC3871572 DOI: 10.1371/journal.pone.0083835] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 11/17/2013] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) has been mainly attributed to white matter (WM) pathology. However, recent evidence indicated the presence of grey matter (GM) lesions. One of the principal mediators of inflammatory processes is interleukin-1β (IL-1β), which is known to play a role in MS pathogenesis. It is unknown whether IL-1β is solely present in WM or also in GM lesions. Using an experimental MS model, we questioned whether IL-1β and the IL-1 receptor antagonist (IL-1ra) are present in GM in addition to affected WM regions. METHODS The expression of IL-1β and IL-1ra in chronic-relapsing EAE (cr-EAE) rats was examined using in situ hybridization, immunohistochemistry and real-time PCR. Rats were sacrificed at the peak of the first disease phase, the trough of the remission phase, and at the peak of the relapse. Histopathological characteristics of CNS lesions were studied using immunohistochemistry for PLP, CD68 and CD3 and Oil-Red O histochemistry. RESULTS IL-1β and IL-ra expression appears to a similar extent in affected GM and WM regions in the brain and spinal cord of cr-EAE rats, particularly in perivascular and periventricular locations. IL-1β and IL-1ra expression was dedicated to macrophages and/or activated microglial cells, at sites of starting demyelination. The time-dependent expression of IL-1β and IL-1ra revealed that within the spinal cord IL-1β and IL-1ra mRNA remained present throughout the disease, whereas in the brain their expression disappeared during the relapse. CONCLUSIONS The appearance of IL-1β expressing cells in GM within the CNS during cr-EAE may explain the occurrence of several clinical deficits present in EAE and MS which cannot be attributed solely to the presence of IL-1β in WM. Endogenously produced IL-1ra seems not capable to counteract IL-1β-induced effects. We put forward that IL-1β may behold promise as a target to address GM, in addition to WM, related pathology in MS.
Collapse
Affiliation(s)
- Marloes Prins
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, The Netherlands
| | - Charlotta Eriksson
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, The Netherlands
| | - Anne Wierinckx
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, The Netherlands
- UNIV UMR1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - John G. J. M. Bol
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, The Netherlands
| | - Rob Binnekade
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, The Netherlands
| | - Fred J. H. Tilders
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, The Netherlands
| | - Anne-Marie Van Dam
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, The Netherlands
| |
Collapse
|
48
|
Mevalonate kinase deficiency and neuroinflammation: balance between apoptosis and pyroptosis. Int J Mol Sci 2013; 14:23274-88. [PMID: 24287904 PMCID: PMC3876043 DOI: 10.3390/ijms141223274] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Revised: 11/08/2013] [Accepted: 11/13/2013] [Indexed: 12/20/2022] Open
Abstract
Mevalonic aciduria, a rare autosomal recessive disease, represents the most severe form of the periodic fever, known as Mevalonate Kinase Deficiency. This disease is caused by the mutation of the MVK gene, which codes for the enzyme mevalonate kinase, along the cholesterol pathway. Mevalonic aciduria patients show recurrent fever episodes with associated inflammatory symptoms, severe neurologic impairments, or death, in early childhood. The typical neurodegeneration occurring in mevalonic aciduria is linked both to the intrinsic apoptosis pathway (caspase-3 and -9), which is triggered by mitochondrial damage, and to pyroptosis (caspase-1). These cell death mechanisms seem to be also related to the assembly of the inflammasome, which may, in turn, activate pro-inflammatory cytokines and chemokines. Thus, this particular molecular platform may play a crucial role in neuroinflammation mechanisms. Nowadays, a specific therapy is still lacking and the pathogenic mechanisms involving neuroinflammation and neuronal dysfunction have not yet been completely understood, making mevalonic aciduria an orphan drug disease. This review aims to analyze the relationship among neuroinflammation, mitochondrial damage, programmed cell death, and neurodegeneration. Targeting inflammation and degeneration in the central nervous system might help identify promising treatment approaches for mevalonic aciduria or other diseases in which these mechanisms are involved.
Collapse
|
49
|
Zhao AP, Dong YF, Liu W, Gu J, Sun XL. Nicorandil inhibits inflammasome activation and Toll-like receptor-4 signal transduction to protect against oxygen-glucose deprivation-induced inflammation in BV-2 cells. CNS Neurosci Ther 2013; 20:147-53. [PMID: 24256503 DOI: 10.1111/cns.12178] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 08/11/2013] [Accepted: 08/12/2013] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Our previous studies have demonstrated adenosine triphosphate-sensitive potassium channel (KATP channel) openers could protect against inflammatory response in brain disease, but little is known about the mechanisms involved in KATP channel openers inhibiting neuroinflammation. METHODS AND RESULTS In the present study, we found that oxygen-glucose deprivation (OGD) resulted in BV-2 cells activation, significantly increased tumor necrosis factor-alpha and interleukin-1beta (IL-1β) levels, accompanied by downregulating Kir6.1 subunit. Pretreatment with nicorandil, a KATP channel opener, could attenuate OGD-induced BV-2 cells activation and inhibit pro-inflammatory factors release. Further study demonstrated that OGD activated Toll-like receptor-4 (TLR4) signaling pathway and NOD-like receptor pyrin domain containing three inflammasome, thereby increased IL-1β production. Pretreatment with nicorandil could reverse the two pathways involved in IL-1β production. CONCLUSIONS Our findings reveal that KATP channel openers could protect against OGD-induced neuroinflammation via inhibiting inflammasome activation and TLR4 signal transduction.
Collapse
Affiliation(s)
- An-Peng Zhao
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | | | | | | | | |
Collapse
|
50
|
Jullienne A, Badaut J. Molecular contributions to neurovascular unit dysfunctions after brain injuries: lessons for target-specific drug development. FUTURE NEUROLOGY 2013; 8:677-689. [PMID: 24489483 PMCID: PMC3904383 DOI: 10.2217/fnl.13.55] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The revised 'expanded' neurovascular unit (eNVU) is a physiological and functional unit encompassing endothelial cells, pericytes, smooth muscle cells, astrocytes and neurons. Ischemic stroke and traumatic brain injury are acute brain injuries directly affecting the eNVU with secondary damage, such as blood-brain barrier (BBB) disruption, edema formation and hypoperfusion. BBB dysfunctions are observed at an early postinjury time point, and are associated with eNVU activation of proteases, such as tissue plasminogen activator and matrix metalloproteinases. BBB opening is accompanied by edema formation using astrocytic AQP4 as a key protein regulating water movement. Finally, nitric oxide dysfunction plays a dual role in association with BBB injury and dysregulation of cerebral blood flow. These mechanisms are discussed including all targets of eNVU encompassing endothelium, glial cells and neurons, as well as larger blood vessels with smooth muscle. In fact, the feeding blood vessels should also be considered to treat stroke and traumatic brain injury. This review underlines the importance of the eNVU in drug development aimed at improving clinical outcome after stroke and traumatic brain injury.
Collapse
Affiliation(s)
- Amandine Jullienne
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Jérôme Badaut
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| |
Collapse
|