ijms-logo

Journal Browser

Journal Browser

Pharmacological Strategies for Neuroinflammation in Brain Injury

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Pharmacology".

Deadline for manuscript submissions: closed (31 October 2022) | Viewed by 17321

Special Issue Editors


E-Mail Website
Guest Editor
Johns Hopkins University School of Medicine, Baltimore, MD, USA
Interests: neuroscience; bone biology; clinical trial; pharmacovigilance
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Department Neurosurgery, University Clinic of Würzburg, Würzburg, Germany
Interests: mechanisms of neuroprotection and -regeneration after brain injury; presynaptic structural plasticity; cell culture; transgenic animals; experimental models of brain trauma; behavioural testing; microscopy and imaging

Special Issue Information

Dear Colleagues,

Brain injury is a widespread and devastating problem that affects all ages. Only incremental improvements in treatment have been made over the past century. Brain injury management lacks effective pharmacological treatment. Persisting neuroinflammation is a significant component of secondary brain injury that can progressively worsen brain damage over time, thus allowing for opportunities to pharmacologically intervene. The management of neuroinflammation may be a promising target for improving patient outcomes. Substantial evidence suggests that therapies targeting the cytokine pathway may limit neuroinflammation. A better understanding of microglia heterogeneity may lead to more effective and precise therapeutic approaches and help to identify biomarkers for brain injury.

The effects of brain injury can be severe, including neurocognitive, physical, and psychosocial impairment. A significant unmet need to develop strategies to avoid long-term damage as a result of brain injury remains. The primary phase of brain injury describes immediate neuronal damage from contusions or oxygen deprivation caused by the global mass effect. The secondary injury occurs later via mechanisms such as reperfusion injury, delayed cortical edema, blood–brain barrier breakdown, and local electrolyte imbalance. These disturbances result in increased reactive oxygen species (ROS), calcium release, glutamate toxicity, lipid peroxidation (LP), and mitochondrial dysfunction, which lead to a vicious positive feedback loop of progressive oxidative stress-mediated neurodegeneration and neuroinflammation. Such a secondary injury may occur in the brain adjacent to the site of the initial injury, yielding an unexpected spread of cellular damage months after post-injury. ROS scavengers and LP product inhibitors have become candidate compounds to treat secondary brain injury. However, there are still no effective treatment options demonstrating improved outcomes in large, multi-center phase III trials. This may be partially attributed to a poor delivery to and an insufficient retention in the brain of the potential therapeutic agents.

Dr. Vijay Kumar
Prof. Dr. Anna-Leena Sirén
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Neuroinflammation in brain injury
  • Oxidative stress in brain injury
  • Blood–brain barrier and brain injury
  • Calcium and brain injury
  • Glutamate and brain injury
  • Lipid peroxidation and brain injury
  • Mitochondrial dysfunction and brain injury
  • Microglia and brain injury
  • Proteotoxicity and brain injury

Published Papers (7 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

16 pages, 22267 KiB  
Article
Multifaceted Benefits of GDF11 Treatment in Spinal Cord Injury: In Vitro and In Vivo Studies
by May-Jywan Tsai, Li-Yu Fay, Dann-Ying Liou, Yi Chen, Ya-Tzu Chen, Meng-Jen Lee, Tsung-Hsi Tu, Wen-Cheng Huang and Henrich Cheng
Int. J. Mol. Sci. 2023, 24(1), 421; https://doi.org/10.3390/ijms24010421 - 27 Dec 2022
Cited by 2 | Viewed by 2068
Abstract
Traumatic spinal cord injury (SCI) initiates a series of cellular and molecular events that include both primary and secondary injury cascades. This secondary cascade provides opportunities for the delivery of therapeutic intervention. Growth differentiation factor 11 (GDF11), a member of the transforming growth [...] Read more.
Traumatic spinal cord injury (SCI) initiates a series of cellular and molecular events that include both primary and secondary injury cascades. This secondary cascade provides opportunities for the delivery of therapeutic intervention. Growth differentiation factor 11 (GDF11), a member of the transforming growth factor-β (TGF-β) superfamily, regulates various biological processes in mammals. The effects of GDF11 in the nervous system were not fully elucidated. Here, we perform extensive in vitro and in vivo studies to unravel the effects of GDF11 on spinal cord after injury. In vitro culture studies showed that GDF11 increased the survival of both neuronal and oligodendroglial cells but decreased microglial cells. In stressed cultures, GDF11 effectively inhibited LPS stimulation and also protected neurons from ischemic damage. Intravenous GDF11 administration to rat after eliciting SCI significantly improved hindlimb functional restoration of SCI rats. Reduced neuronal connectivity was evident at 6 weeks post-injury and these deficits were markedly attenuated by GDF11 treatment. Furthermore, SCI-associated oligodendroglial alteration were more preserved by GDF11 treatment. Taken together, GDF11 infusion via intravenous route to SCI rats is beneficial, facilitating its therapeutic application in the future. Full article
(This article belongs to the Special Issue Pharmacological Strategies for Neuroinflammation in Brain Injury)
Show Figures

Figure 1

19 pages, 4042 KiB  
Article
Novel Hydroxypyridine Compound Protects Brain Cells against Ischemic Damage In Vitro and In Vivo
by Ekaterina Blinova, Egor Turovsky, Elena Eliseikina, Alexandra Igrunkova, Elena Semeleva, Grigorii Golodnev, Rita Termulaeva, Olga Vasilkina, Sofia Skachilova, Yan Mazov, Kirill Zhandarov, Ekaterina Simakina, Konstantin Belanov, Saveliy Zalogin and Dmitrii Blinov
Int. J. Mol. Sci. 2022, 23(21), 12953; https://doi.org/10.3390/ijms232112953 - 26 Oct 2022
Cited by 2 | Viewed by 1595
Abstract
A non-surgical pharmacological approach to control cellular vitality and functionality during ischemic and/or reperfusion-induced phases of strokes remains extremely important. The synthesis of 2-ethyl-6-methyl-3-hydroxypyridinium gammalactone-2,3-dehydro-L-gulonate (3-EA) was performed using a topochemical reaction. The cell-protective effects of 3-EA were studied on a model of [...] Read more.
A non-surgical pharmacological approach to control cellular vitality and functionality during ischemic and/or reperfusion-induced phases of strokes remains extremely important. The synthesis of 2-ethyl-6-methyl-3-hydroxypyridinium gammalactone-2,3-dehydro-L-gulonate (3-EA) was performed using a topochemical reaction. The cell-protective effects of 3-EA were studied on a model of glutamate excitotoxicity (GluTox) and glucose-oxygen deprivation (OGD) in a culture of NMRI mice cortical cells. Ca2+ dynamics was studied using fluorescent bioimaging and a Fura-2 probe, cell viability was assessed using cytochemical staining with propidium iodide, and gene expression was assessed by a real-time polymerase chain reaction. The compound anti-ischemic efficacy in vivo was evaluated on a model of irreversible middle cerebral artery (MCA) occlusion in Sprague-Dawley male rats. Brain morphological changes and antioxidant capacity were assessed one week after the pathology onset. The severity of neurological disorder was evaluated dynamically. 3-EA suppressed cortical cell death in a dose-dependent manner under the excitotoxic effect of glutamate and ischemia/reoxygenation. Pre-incubation of cerebral cortex cells with 10–100 µM 3-EA led to significant stagnation in Ca2+ concentration in a cytosol ([Ca2+]i) of neurons and astrocytes suffering GluTox and OGD. Decreasing intracellular Ca2+ and establishing a lower [Ca2+]i baseline inhibited necrotic cell death in an acute experiment. The mechanism of 3-EA cytoprotective action involved changes in the baseline and ischemia/reoxygenation-induced expression of genes encoding anti-apoptotic proteins and proteins of the oxidative status; this led to inhibition of the late irreversible stages of apoptosis. Incubation of brain cortex cells with 3-EA induced an overexpression of the anti-apoptotic genes BCL-2, STAT3, and SOCS3, whereas the expression of genes regulating necrosis and inflammation (TRAIL, MLKL, Cas-1, Cas-3, IL-1β and TNFa) were suppressed. 3-EA 18.0 mg/kg intravenous daily administration for 7 days following MCA occlusion preserved rats’ cortex neuron population, decreased the severity of neurological deficit, and spared antioxidant capacity of damaged tissues. 3-EA demonstrated proven short-term anti-ischemic activity in vivo and in vitro, which can be associated with antioxidant activity and the ability to target necrotic and apoptotic death. The compound may be considered a potential neuroprotective molecule for further pre-clinical investigation. Full article
(This article belongs to the Special Issue Pharmacological Strategies for Neuroinflammation in Brain Injury)
Show Figures

Figure 1

21 pages, 6361 KiB  
Article
Lupeol Treatment Attenuates Activation of Glial Cells and Oxidative-Stress-Mediated Neuropathology in Mouse Model of Traumatic Brain Injury
by Riaz Ahmad, Amjad Khan, Inayat Ur Rehman, Hyeon Jin Lee, Ibrahim Khan and Myeong Ok Kim
Int. J. Mol. Sci. 2022, 23(11), 6086; https://doi.org/10.3390/ijms23116086 - 29 May 2022
Cited by 12 | Viewed by 3239
Abstract
Traumatic brain injury (TBI) signifies a major cause of death and disability. TBI causes central nervous system (CNS) damage under a variety of mechanisms, including protein aggregation, mitochondrial dysfunction, oxidative stress, and neuroinflammation. Astrocytes and microglia, cells of the CNS, are considered the [...] Read more.
Traumatic brain injury (TBI) signifies a major cause of death and disability. TBI causes central nervous system (CNS) damage under a variety of mechanisms, including protein aggregation, mitochondrial dysfunction, oxidative stress, and neuroinflammation. Astrocytes and microglia, cells of the CNS, are considered the key players in initiating an inflammatory response after injury. Several evidence suggests that activation of astrocytes/microglia and ROS/LPO have the potential to cause more harmful effects in the pathological processes following traumatic brain injury (TBI). Previous studies have established that lupeol provides neuroprotection through modulation of inflammation, oxidative stress, and apoptosis in Aβ and LPS model and neurodegenerative disease. However, the effects of lupeol on apoptosis caused by inflammation and oxidative stress in TBI have not yet been investigated. Therefore, we explored the role of Lupeol on antiapoptosis, anti-inflammatory, and antioxidative stress and its potential mechanism following TBI. In these experiments, adult male mice were randomly divided into four groups: control, TBI, TBI+ Lupeol, and Sham group. Western blotting, immunofluorescence staining, and ROS/LPO assays were performed to investigate the role of lupeol against neuroinflammation, oxidative stress, and apoptosis. Lupeol treatment reversed TBI-induced behavioral and memory disturbances. Lupeol attenuated TBI-induced generation of reactive oxygen species/lipid per oxidation (ROS/LPO) and improved the antioxidant protein level, such as nuclear factor erythroid 2-related factor 2 (Nrf2) and heme-oxygenase 1 (HO-1) in the mouse brain. Similarly, our results indicated that lupeol treatment inhibited glial cell activation, p-NF-κB, and downstream signaling molecules, such as TNF-α, COX-2, and IL-1β, in the mouse cortex and hippocampus. Moreover, lupeol treatment also inhibited mitochondrial apoptotic signaling molecules, such as caspase-3, Bax, cytochrome-C, and reversed deregulated Bcl2 in TBI-treated mice. Overall, our study demonstrated that lupeol inhibits the activation of astrocytes/microglia and ROS/LPO that lead to oxidative stress, neuroinflammation, and apoptosis followed by TBI. Full article
(This article belongs to the Special Issue Pharmacological Strategies for Neuroinflammation in Brain Injury)
Show Figures

Figure 1

18 pages, 6112 KiB  
Article
Anti-Inflammatory Effect of IKK-Activated GSK-3β Inhibitory Peptide Prevented Nigrostriatal Neurodegeneration in the Rodent Model of Parkinson’s Disease
by Seulah Lee, Dong Geun Hong, Seonguk Yang, Jaehoon Kim, Minwoo Baek, Seoyeong Kim, Dinakaran Thirumalai, Hae Young Chung, Seung-Cheol Chang and Jaewon Lee
Int. J. Mol. Sci. 2022, 23(2), 998; https://doi.org/10.3390/ijms23020998 - 17 Jan 2022
Cited by 7 | Viewed by 2941
Abstract
Parkinson’s disease (PD) is a progressive movement disorder caused by nigrostriatal neurodegeneration. Since chronically activated neuroinflammation accelerates neurodegeneration in PD, we considered that modulating chronic neuroinflammatory response might provide a novel therapeutic approach. Glycogen synthase kinase 3 (GSK-3) is a multifunctional serine/threonine protein [...] Read more.
Parkinson’s disease (PD) is a progressive movement disorder caused by nigrostriatal neurodegeneration. Since chronically activated neuroinflammation accelerates neurodegeneration in PD, we considered that modulating chronic neuroinflammatory response might provide a novel therapeutic approach. Glycogen synthase kinase 3 (GSK-3) is a multifunctional serine/threonine protein kinase with two isoforms, GSK-3α and GSK-3β, and GSK-3β plays crucial roles in inflammatory response, which include microglial migration and peripheral immune cell activation. GSK-3β inhibitory peptide (IAGIP) is specifically activated by activated inhibitory kappa B kinase (IKK), and its therapeutic effects have been demonstrated in a mouse model of colitis. Here, we investigated whether the anti-inflammatory effects of IAGIP prevent neurodegeneration in the rodent model of PD. IAGIP significantly reduced MPP+-induced astrocyte activation and inflammatory response in primary astrocytes without affecting the phosphorylations of ERK or JNK. In addition, IAGIP inhibited LPS-induced cell migration and p65 activation in BV-2 microglial cells. In vivo study using an MPTP-induced mouse model of PD revealed that intravenous IAGIP effectively prevented motor dysfunction and nigrostriatal neurodegeneration. Our findings suggest that IAGIP has a curative potential in PD models and could offer new therapeutic possibilities for targeting PD. Full article
(This article belongs to the Special Issue Pharmacological Strategies for Neuroinflammation in Brain Injury)
Show Figures

Figure 1

10 pages, 1057 KiB  
Article
FosL1 Is a Novel Target of Levetiracetam for Suppressing the Microglial Inflammatory Reaction
by Kouji Niidome, Ruri Taniguchi, Takeshi Yamazaki, Mayumi Tsuji, Kouichi Itoh and Yasuhiro Ishihara
Int. J. Mol. Sci. 2021, 22(20), 10962; https://doi.org/10.3390/ijms222010962 - 11 Oct 2021
Cited by 9 | Viewed by 2443
Abstract
We previously showed that the antiepileptic drug levetiracetam (LEV) inhibits microglial activation, but the mechanism remains unclear. The purpose of this study was to identify the target of LEV in microglial activity suppression. The mouse microglial BV-2 cell line, cultured in a ramified [...] Read more.
We previously showed that the antiepileptic drug levetiracetam (LEV) inhibits microglial activation, but the mechanism remains unclear. The purpose of this study was to identify the target of LEV in microglial activity suppression. The mouse microglial BV-2 cell line, cultured in a ramified form, was pretreated with LEV and then treated with lipopolysaccharide (LPS). A comprehensive analysis of LEV targets was performed by cap analysis gene expression sequencing using BV-2 cells, indicating the transcription factors BATF, Nrf-2, FosL1 (Fra1), MAFF, and Spic as candidates. LPS increased AP-1 and Spic transcriptional activity, and LEV only suppressed AP-1 activity. FosL1, MAFF, and Spic mRNA levels were increased by LPS, and LEV only attenuated FosL1 mRNA expression, suggesting FosL1 as an LEV target. FosL1 protein levels were increased by LPS treatment and decreased by LEV pretreatment, similar to FosL1 mRNA levels. The FosL1 siRNA clearly suppressed the expression of TNFα and IL-1β. Pilocarpine-induced status epilepticus increased hippocampus FosL1 expression, along with inflammation. LEV treatment significantly suppressed FosL1 expression. Together, LEV reduces FosL1 expression and AP-1 activity in activated microglia, thereby suppressing neuroinflammation. LEV might be a candidate for the treatment of several neurological diseases involving microglial activation. Full article
(This article belongs to the Special Issue Pharmacological Strategies for Neuroinflammation in Brain Injury)
Show Figures

Figure 1

Review

Jump to: Research

16 pages, 1024 KiB  
Review
Cerebrospinal Fluid Biomarkers for Diagnosis and the Prognostication of Acute Ischemic Stroke: A Systematic Review
by Anant Naik, Olufunmilola Adeleye, Stefan W. Koester, Ethan A. Winkler, Joelle N. Hartke, Katherine Karahalios, Sandra Mihaljevic, Anupama Rani, Sudhanshu Raikwar, Jarrod D. Rulney, Shashvat M. Desai, Lea Scherschinski, Andrew F. Ducruet, Felipe C. Albuquerque, Michael T. Lawton, Joshua S. Catapano, Ashutosh P. Jadhav and Ruchira M. Jha
Int. J. Mol. Sci. 2023, 24(13), 10902; https://doi.org/10.3390/ijms241310902 - 30 Jun 2023
Cited by 3 | Viewed by 1934
Abstract
Despite the high incidence and burden of stroke, biological biomarkers are not used routinely in clinical practice to diagnose, determine progression, or prognosticate outcomes of acute ischemic stroke (AIS). Because of its direct interface with neural tissue, cerebrospinal fluid (CSF) is a potentially [...] Read more.
Despite the high incidence and burden of stroke, biological biomarkers are not used routinely in clinical practice to diagnose, determine progression, or prognosticate outcomes of acute ischemic stroke (AIS). Because of its direct interface with neural tissue, cerebrospinal fluid (CSF) is a potentially valuable source for biomarker development. This systematic review was conducted using three databases. All trials investigating clinical and preclinical models for CSF biomarkers for AIS diagnosis, prognostication, and severity grading were included, yielding 22 human trials and five animal studies for analysis. In total, 21 biomarkers and other multiomic proteomic markers were identified. S100B, inflammatory markers (including tumor necrosis factor-alpha and interleukin 6), and free fatty acids were the most frequently studied biomarkers. The review showed that CSF is an effective medium for biomarker acquisition for AIS. Although CSF is not routinely clinically obtained, a potential benefit of CSF studies is identifying valuable biomarkers from the pathophysiologic microenvironment that ultimately inform optimization of targeted low-abundance assays from peripheral biofluid samples (e.g., plasma). Several important catabolic and anabolic markers can serve as effective measures of diagnosis, etiology identification, prognostication, and severity grading. Trials with large cohorts studying the efficacy of biomarkers in altering clinical management are still needed. Full article
(This article belongs to the Special Issue Pharmacological Strategies for Neuroinflammation in Brain Injury)
Show Figures

Figure 1

17 pages, 5890 KiB  
Review
Anti-Inflammatory Drug Therapy in Chronic Subdural Hematoma: A Systematic Review and Meta-Analysis of Prospective Randomized, Double-Blind and Placebo-Controlled Trials
by Martin Vychopen, Erdem Güresir and Johannes Wach
Int. J. Mol. Sci. 2022, 23(24), 16198; https://doi.org/10.3390/ijms232416198 - 19 Dec 2022
Cited by 3 | Viewed by 1875
Abstract
Althoughanti-inflammatory drug therapy has been identified as potentially beneficial for patients suffering from chronic subdural hematoma (cSDH), contemporary literature presents contradictory results. In this meta-analysis, we aimed to investigate the impact of anti-inflammatory drug therapy on mortality and outcome. We searched for eligible [...] Read more.
Althoughanti-inflammatory drug therapy has been identified as potentially beneficial for patients suffering from chronic subdural hematoma (cSDH), contemporary literature presents contradictory results. In this meta-analysis, we aimed to investigate the impact of anti-inflammatory drug therapy on mortality and outcome. We searched for eligible randomized, placebo-controlled prospective trials (RTCs) on PubMed, Embase and Medline until July 2022. From 97 initially identified articles, five RTCs met the criteria and were included in our meta-analysis. Our results illustrate significantly lower rates of recurrent cSDH (OR: 0.35; 95% CI: 0.21–0.58, p = 0.0001) in patients undergoing anti-inflammatory therapy. In the subgroup of patients undergoing primary conservative treatment, anti-inflammatory therapy was associated with lower rates of “switch to surgery” cases (OR: 0.30; 95% CI: 0.14–0.63, p = 0.002). Despite these findings, anti-inflammatory drugs seemed to be associated with higher mortality rates in patients undergoing surgery (OR: 1.76; 95% CI: 1.03–3.01, p = 0.04), although in the case of primary conservative treatment, no effect on mortality has been observed (OR: 2.45; 95% CI: 0.35–17.15, p = 0.37). Further multicentric prospective randomized trials are needed to evaluate anti-inflammatory drugs as potentially suitable therapy for asymptomatic patients with cSDH to avoid the necessity of surgical hematoma evacuation on what are predominantly elderly, vulnerable, patients. Full article
(This article belongs to the Special Issue Pharmacological Strategies for Neuroinflammation in Brain Injury)
Show Figures

Figure 1

Back to TopTop