1
|
Emre Aydıngöz S, Teimoori A, Orhan HG, Demirtaş E, Zeynalova N. A meta-analysis of animal studies evaluating the effect of hydrogen sulfide on ischemic stroke: is the preclinical evidence sufficient to move forward? NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9533-9548. [PMID: 39017715 PMCID: PMC11582254 DOI: 10.1007/s00210-024-03291-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024]
Abstract
Hydrogen sulfide (H2S) is a gasotransmitter that has been studied for its potential therapeutic effects, including its role in the pathophysiology and treatment of stroke. This systematic review and meta-analysis aimed to determine the sufficiency of overall preclinical evidence to guide the initiation of clinical stroke trials with H2S and provide tailored recommendations for their design. PubMed, Web of Science, Scopus, EMBASE, and MEDLINE were searched for studies evaluating the effect of any H2S donor on in vivo animal models of regional ischemic stroke, and 34 publications were identified. Pooling of the effect sizes using the random-effect model revealed that H2S decreased the infarct area by 34.5% (95% confidence interval (CI) 28.2-40.8%, p < 0.0001), with substantial variability among the studies (I2 = 89.8%). H2S also caused a 37.9% reduction in the neurological deficit score (95% CI 29.0-46.8%, p < 0.0001, I2 = 63.8%) and in the brain water content (3.2%, 95% CI 1.4-4.9%, p = 0.0014, I2 = 94.6%). Overall, the studies had a high risk of bias and low quality of evidence (median quality score 5/15, interquartile range 4-9). The majority of the included studies had a "high" or "unclear" risk of bias, and none of the studies overall had a "low" risk. In conclusion, H2S significantly improves structural and functional outcomes in in vivo animal models of ischemic stroke. However, the level of evidence from preclinical studies is not sufficient to proceed to clinical trials due to the low external validity, high risk of bias, and variable design of existing animal studies.
Collapse
Affiliation(s)
- Selda Emre Aydıngöz
- Department of Medical Pharmacology, Başkent University Faculty of Medicine, Ankara, Turkey.
| | - Ariyan Teimoori
- Department of Medical Pharmacology, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Halit Güner Orhan
- Department of Medical Pharmacology, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Elif Demirtaş
- Department of Medical Pharmacology, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Nargız Zeynalova
- Department of Medical Pharmacology, Başkent University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
2
|
Benkő S, Dénes Á. Microglial Inflammatory Mechanisms in Stroke: The Jury Is Still Out. Neuroscience 2024; 550:43-52. [PMID: 38364965 DOI: 10.1016/j.neuroscience.2024.02.007] [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: 01/04/2024] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 02/18/2024]
Abstract
Microglia represent the main immune cell population in the CNS with unique homeostatic roles and contribution to broad neurological conditions. Stroke is associated with marked changes in microglial phenotypes and induction of inflammatory responses, which emerge as key modulators of brain injury, neurological outcome and regeneration. However, due to the limited availability of functional studies with selective targeting of microglia and microglia-related inflammatory pathways in stroke, the vast majority of observations remain correlative and controversial. Because extensive review articles discussing the role of inflammatory mechanisms in different forms of acute brain injury are available, here we focus on some specific pathways that appear to be important for stroke pathophysiology with assumed contribution by microglia. While the growing toolkit for microglia manipulation increasingly allows targeting inflammatory pathways in a cell-specific manner, reconsideration of some effects devoted to microglia may also be required. This may particularly concern the interpretation of inflammatory mechanisms that emerge in response to stroke as a form of sterile injury and change markedly in chronic inflammation and common stroke comorbidities.
Collapse
Affiliation(s)
- Szilvia Benkő
- Laboratory of Inflammation-Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Ádám Dénes
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest H-1083, Hungary.
| |
Collapse
|
3
|
Pérez-Mato M, López-Arias E, Bugallo-Casal A, Correa-Paz C, Arias S, Rodríguez-Yáñez M, Santamaría-Cadavid M, Campos F. New Perspectives in Neuroprotection for Ischemic Stroke. Neuroscience 2024; 550:30-42. [PMID: 38387732 DOI: 10.1016/j.neuroscience.2024.02.017] [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: 11/01/2023] [Revised: 02/12/2024] [Accepted: 02/16/2024] [Indexed: 02/24/2024]
Abstract
The constant failure of new neuroprotective therapies for ischemic stroke has partially halted the search for new therapies in recent years, mainly because of the high investment risk required to develop a new treatment for a complex pathology, such as stroke, with a narrow intervention window and associated comorbidities. However, owing to recent progress in understanding the stroke pathophysiology, improvement in patient care in stroke units, development of new imaging techniques, search for new biomarkers for early diagnosis, and increasingly widespread use of mechanical recanalization therapies, new opportunities have opened for the study of neuroprotection. This review summarizes the main protective agents currently in use, some of which are already in the clinical evaluation phase. It also includes an analysis of how recanalization therapies, new imaging techniques, and biomarkers have improved their efficacy.
Collapse
Affiliation(s)
- María Pérez-Mato
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Esteban López-Arias
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Ana Bugallo-Casal
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Clara Correa-Paz
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Susana Arias
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, 15706 Santiago de Compostela, Spain
| | - Manuel Rodríguez-Yáñez
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, 15706 Santiago de Compostela, Spain
| | - María Santamaría-Cadavid
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, 15706 Santiago de Compostela, Spain
| | - Francisco Campos
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
4
|
Cliteur MP, van der Kolk AG, Hannink G, Hofmeijer J, Jolink WMT, Klijn CJM, Schreuder FHBM. Anakinra in cerebral haemorrhage to target secondary injury resulting from neuroinflammation (ACTION): Study protocol of a phase II randomised clinical trial. Eur Stroke J 2024; 9:265-273. [PMID: 37713268 PMCID: PMC10916813 DOI: 10.1177/23969873231200686] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/10/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND Inflammation plays a vital role in the development of secondary brain injury after spontaneous intracerebral haemorrhage (ICH). Interleukin-1 beta is an early pro-inflammatory cytokine and a potential therapeutic target. AIM To determine the effect of treatment with recombinant human interleukin-1 receptor antagonist anakinra on perihematomal oedema (PHO) formation in patients with spontaneous ICH compared to standard medical management, and investigate whether this effect is dose-dependent. METHODS ACTION is a phase-II, prospective, randomised, three-armed (1:1:1) trial with open-label treatment and blinded end-point assessment (PROBE) at three hospitals in The Netherlands. We will include 75 patients with a supratentorial spontaneous ICH admitted within 8 h after symptom onset. Participants will receive anakinra in a high dose (loading dose 500 mg intravenously, followed by infusion with 2 mg/kg/h over 72 h; n = 25) or in a low dose (loading dose 100 mg subcutaneously, followed by 100 mg subcutaneous twice daily for 72 h; n = 25), plus standard care. The control group (n = 25) will receive standard medical management. OUTCOMES Primary outcome is PHO, measured as oedema extension distance on MRI at day 7 ± 1. Secondary outcomes include the safety profile of anakinra, the effect of anakinra on serum inflammation markers, MRI measures of blood brain barrier integrity, and functional outcome at 90 ± 7 days. DISCUSSION The ACTION trial will provide insight into whether targeting interleukin-1 beta in the early time window after ICH onset could ameliorate secondary brain injury. This may contribute to the development of new treatment options to improve clinical outcome after ICH.
Collapse
Affiliation(s)
- MP Cliteur
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - AG van der Kolk
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - G Hannink
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - J Hofmeijer
- Department of Neurology, Rijnstate Hospital, Arnhem, The Netherlands
- Department of Clinical Neurophysiology, University of Twente, Enschede, The Netherlands
| | - WMT Jolink
- Department of Neurology, Isala Hospital, Zwolle, The Netherlands
| | - CJM Klijn
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - FHBM Schreuder
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
5
|
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
|
6
|
Wilson E, Currie G, Macleod M, Kind P, Sena ES. Genetically modified animals as models of neurodevelopmental conditions: A review of systematic review reporting quality. Brain Neurosci Adv 2024; 8:23982128241287279. [PMID: 39431203 PMCID: PMC11489925 DOI: 10.1177/23982128241287279] [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: 07/12/2024] [Accepted: 09/05/2024] [Indexed: 10/22/2024] Open
Abstract
Using genetically modified animals to model neurodevelopmental conditions helps better our understanding of biology underlying these conditions. Animal research has unique characteristics not shared with clinical research, meaning systematic review methods must be adapted to this context. We aim to evaluate the quantity, characteristics, and reporting quality of systematic reviews which synthesise research using genetically modified animals to model neurodevelopmental conditions. On 23 January 2023, we searched PubMed, Embase, and the Web of Science Core Collection to identify systematic reviews of genetic neurodevelopmental condition animal research where the modified gene was one in a list of 102 genes associated with neurodevelopmental conditions identified through large-scale exome sequencing or Fmr1, Mecp2, or Ube3a. Two independent reviewers screened studies based on full text and assessed the reporting quality of relevant reviews using an adapted version of the PRISMA checklist (PRISMA-Pre). Twelve review publications met our criteria. We found mixed levels of reporting: items such as identifying the publication as a systematic review in the title, search strategies, and funding sources being well reported, and others such as protocol registration and data sharing less well reported. We also identified 19 review registrations via PROSPERO, most of which remain unpublished after their anticipated end dates. Systematic reviews are limited by lack of reporting. Increased awareness of reporting guidelines may help authors increase the transparency and reproducibility, and therefore the reliability, of their systematic reviews.
Collapse
Affiliation(s)
- Emma Wilson
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, The University of Edinburgh, Edinburgh, UK
| | - Gillian Currie
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Malcolm Macleod
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Peter Kind
- Simons Initiative for the Developing Brain, The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- Patrick Wild Centre for Autism Research, The University of Edinburgh, Edinburgh, UK
| | - Emily S. Sena
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
7
|
López-Morales MA, Castelló-Ruiz M, Burguete MC, Hervás D, Pérez-Pinzón MA, Salom JB. Effect and mechanisms of resveratrol in animal models of ischemic stroke: A systematic review and Bayesian meta-analysis. J Cereb Blood Flow Metab 2023; 43:2013-2028. [PMID: 37802493 PMCID: PMC10925864 DOI: 10.1177/0271678x231206236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 08/03/2023] [Accepted: 09/20/2023] [Indexed: 10/10/2023]
Abstract
Resveratrol (RSV) holds promise as cerebroprotective treatment in cerebral ischemia. This systematic review aims to assess the effects and mechanisms of RSV in animal models of ischemic stroke. We searched Medline, Embase and Web of Science to identify 75 and 57 eligible rodent studies for qualitative and quantitative syntheses, respectively. Range of evidence met 10 of 13 STAIR criteria. Median (Q1, Q3) quality score was 7 (5, 8) on the CAMARADES 15-item checklist. Bayesian meta-analysis showed SMD estimates (95% CI) favoring RSV: infarct size (-1.72 [-2.03; -1.41]), edema size (-1.61 [-2.24; -0.98]), BBB impairment (-1.85 [-2.54; -1.19]), neurofunctional impairment (-1.60 [-1.92; -1.29]), and motor performance (1.39 [0.64; 2.08]); and less probably neuronal survival (0.63 [-1.40; 2.48]) and apoptosis (-0.96 [-2.87; 1.02]). Species (rat vs mouse) was associated to a larger benefit. Sensitivity analyses confirmed robustness of the estimates. Reduction of oxidative stress, inflammation, and apoptosis underlie these effects. Our results quantitatively state the beneficial effects of RSV on structural and functional outcomes in rodent stroke models, update the evidence on the mechanisms of action, and provide an exhaustive list of targeted signaling pathways. Current evidence highlights the need for conducting further high-quality preclinical research to better inform clinical research.
Collapse
Affiliation(s)
- Mikahela A López-Morales
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - María Castelló-Ruiz
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, Valencia, Spain
| | - María C Burguete
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - David Hervás
- Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain
| | - Miguel A Pérez-Pinzón
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Department of Neurology, Miller School of Medicine, University of Miami, Miami, USA
| | - Juan B Salom
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| |
Collapse
|
8
|
Cai D, Fraunfelder M, Fujise K, Chen SY. ADAR1 exacerbates ischemic brain injury via astrocyte-mediated neuron apoptosis. Redox Biol 2023; 67:102903. [PMID: 37801857 PMCID: PMC10570147 DOI: 10.1016/j.redox.2023.102903] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/08/2023] Open
Abstract
Astrocytes affect stroke outcomes by acquiring functionally dominant phenotypes. Understanding molecular mechanisms dictating astrocyte functional status after brain ischemia/reperfusion may reveal new therapeutic strategies. Adenosine deaminase acting on RNA (ADAR1), an RNA editing enzyme, is not normally expressed in astrocytes, but highly induced in astrocytes in ischemic stroke lesions. The expression of ADAR1 steeply increased from day 1 to day 7 after middle cerebral artery occlusion (MCAO) for 1 h followed by reperfusion. ADAR1 deficiency markedly ameliorated the volume of the cerebral infarction and neurological deficits as shown by the rotarod and cylinder tests, which was due to the reduction of the numbers of activated astrocytes and microglia. Surprisingly, ADAR1 was mainly expressed in astrocytes while only marginally in microglia. In primary cultured astrocytes, ADAR1 promoted astrocyte proliferation via phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Furthermore, ADAR1 deficiency inhibited brain cell apoptosis in mice with MCAO as well as in activated astrocyte-conditioned medium-induced neurons in vitro. It appeared that ADAR1 induces neuron apoptosis by secretion of IL-1β, IL-6 and TNF-α from astrocytes through the production of reactive oxygen species. These results indicated that ADAR1 is a novel regulator promoting the proliferation of the activated astrocytes following ischemic stroke, which produce various inflammatory cytokines, leading to neuron apoptosis and worsened ischemic stroke outcome.
Collapse
Affiliation(s)
- Dunpeng Cai
- Departments of Surgery, University of Missouri School of Medicine, Columbia, MO, USA
| | - Mikayla Fraunfelder
- Departments of Surgery, University of Missouri School of Medicine, Columbia, MO, USA
| | - Ken Fujise
- Harborview Medical Center, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Shi-You Chen
- Departments of Surgery, University of Missouri School of Medicine, Columbia, MO, USA; The Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.
| |
Collapse
|
9
|
Lima TS. Beyond an inflammatory mediator: Interleukin-1 in neurophysiology. Exp Physiol 2023; 108:917-924. [PMID: 37031383 PMCID: PMC10988528 DOI: 10.1113/ep090780] [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: 01/18/2023] [Accepted: 03/20/2023] [Indexed: 04/10/2023]
Abstract
NEW FINDINGS What is the topic of this review? This review focuses on the physiological role of the cytokine interleukin-1β in the CNS. What advances does it highlight? Traditionally, interleukin-1β is known as a key mediator of inflammation and immunity. This review highlights the more recent findings describing how interleukin-1β signalling is required to maintain homeostasis in the CNS. ABSTRACT Since its discovery in the early 1940s, the interleukin-1 (IL-1) cytokine family has been associated primarily with acute and chronic inflammation. The family member IL-1β is produced by different leucocytes, endothelial cells and epithelial cells. This cytokine has been characterized as a key modulator of inflammation and innate immunity because it induces the transcription of several downstream inflammatory genes. More recently, several groups have demonstrated that IL-1β production is also required to maintain homeostasis in several organ systems. This review focuses on providing an overview of the more recently characterized role of IL-1β in the physiology of the CNS. So far, IL-1β signalling has been implicated in neuronal survival, neurite growth, synaptic pruning, synaptic transmission, neuroplasticity and neuroendocrine functions.
Collapse
Affiliation(s)
- Tatiane S. Lima
- Department of Biological SciencesCalifornia State Polytechnic UniversityPomonaCaliforniaUSA
| |
Collapse
|
10
|
Qureshi AI, Akhtar IN, Ma X, Lodhi A, Bhatti I, Beall J, Broderick JP, Cassarly CN, Martin RH, Sharma R, Thakkar M, Suarez JI. Effect of Cilostazol in Animal Models of Cerebral Ischemia and Subarachnoid Hemorrhage: A Systematic Review and Meta-Analysis. Neurocrit Care 2023; 38:698-713. [PMID: 36450971 DOI: 10.1007/s12028-022-01637-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/27/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND Cilostazol, a phosphodiesterase III inhibitor, appears to be a promising agent for preventing cerebral ischemia in patients with aneurysmal subarachnoid hemorrhage. Here, the authors perform a systematic review and meta-analysis to quantitatively assess the effects of cilostazol on brain structural and functional outcomes in animal models of cerebral ischemia and subarachnoid hemorrhage-induced cerebral vasospasm. METHODS By using the PRISMA guidelines, a search of the PubMed, Scopus, and Web of Science was conducted to identify relevant studies. Study quality of each included study for both systematic reviews were scored by using an adapted 15-item checklist from the Collaborative Approach to Meta-Analysis of Animal Data from Experimental Studies. We calculated a standardized mean difference as effect size for each comparison. For each outcome, comparisons were combined by using random-effects modeling to account for heterogeneity, with a restricted maximum likelihood estimate of between-study variance. RESULTS A total of 22 (median [Q1, Q3] quality score of 7 [5, 8]) and 6 (median [Q1, Q3] quality score of 6 [6, 6]) studies were identified for cerebral ischemia and subarachnoid hemorrhage-induced cerebral vasospasm, respectively. Cilostazol significantly reduced the infarct volume in cerebral ischemia models with a pooled standardized mean difference estimate of - 0.88 (95% confidence interval [CI] [- 1.07 to - 0.70], p < 0.0001). Cilostazol significantly reduced neurofunctional deficits in cerebral ischemia models with a pooled standardized mean difference estimate of - 0.66 (95% CI [- 1.06 to - 0.28], p < 0.0001). Cilostazol significantly improved the basilar artery diameter in subarachnoid hemorrhage-induced cerebral vasospasm with a pooled standardized mean difference estimate of 2.30 (95% CI [0.94 to 3.67], p = 0.001). Cilostazol also significantly improved the basilar artery cross-section area with a pooled standardized mean estimate of 1.88 (95% CI [0.33 to 3.43], p < 0.05). Overall, there was between-study heterogeneity and asymmetry in the funnel plot observed in all comparisons. CONCLUSIONS Published animal data support the overall efficacy of cilostazol in reducing infarct volume and neurofunctional deficits in cerebral ischemia models and cerebral vasospasm in subarachnoid hemorrhage models.
Collapse
Affiliation(s)
- Adnan I Qureshi
- Department of Neurology, University of Missouri, Columbia, MO, USA.
- Zeenat Qureshi Stroke Institute, St. Cloud, MN, USA.
| | - Iqra N Akhtar
- Department of Neurology, University of Missouri, Columbia, MO, USA
- Zeenat Qureshi Stroke Institute, St. Cloud, MN, USA
| | - Xiaoyu Ma
- Department of Neurology, University of Missouri, Columbia, MO, USA
- Zeenat Qureshi Stroke Institute, St. Cloud, MN, USA
| | - Abdullah Lodhi
- Department of Neurology, University of Missouri, Columbia, MO, USA
- Zeenat Qureshi Stroke Institute, St. Cloud, MN, USA
| | - Ibrahim Bhatti
- Department of Neurology, University of Missouri, Columbia, MO, USA
- Zeenat Qureshi Stroke Institute, St. Cloud, MN, USA
| | - Jonathan Beall
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | | | - Christy N Cassarly
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Renee H Martin
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Rishi Sharma
- Department of Neurology, University of Missouri, Columbia, MO, USA
| | - Mahesh Thakkar
- Department of Neurology, University of Missouri, Columbia, MO, USA
| | - Jose I Suarez
- Division of Neurosciences Critical Care, Departments of Anesthesiology and Critical Care Medicine, Neurology, and Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
11
|
Advanced methods and implementations for the meta-analyses of animal models: Current practices and future recommendations. Neurosci Biobehav Rev 2023; 146:105016. [PMID: 36566804 DOI: 10.1016/j.neubiorev.2022.105016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Meta-analytic techniques have been widely used to synthesize data from animal models of human diseases and conditions, but these analyses often face two statistical challenges due to complex nature of animal data (e.g., multiple effect sizes and multiple species): statistical dependency and confounding heterogeneity. These challenges can lead to unreliable and less informative evidence, which hinders the translation of findings from animal to human studies. We present a literature survey of meta-analysis using animal models (animal meta-analysis), showing that these issues are not adequately addressed in current practice. To address these challenges, we propose a meta-analytic framework based on multilevel (linear mixed-effects) models. Through conceptualization, formulations, and worked examples, we illustrate how this framework can appropriately address these issues while allowing for testing new questions. Additionally, we introduce other advanced techniques such as multivariate models, robust variance estimation, and meta-analysis of emergent effect sizes, which can deliver robust inferences and novel biological insights. We also provide a tutorial with annotated R code to demonstrate the implementation of these techniques.
Collapse
|
12
|
Fomicheva EE, Shanin SN, Filatenkova TA, Novikova NS, Dyatlova AS, Ishchenko AM, Serebryanaya NB. Correction of Behavioral Disorders and State of Microglia with Recombinant IL-1 Receptor Antagonist in Experimental Traumatic Brain Injury. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022050258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
13
|
Candelario-Jalil E, Dijkhuizen RM, Magnus T. Neuroinflammation, Stroke, Blood-Brain Barrier Dysfunction, and Imaging Modalities. Stroke 2022; 53:1473-1486. [PMID: 35387495 PMCID: PMC9038693 DOI: 10.1161/strokeaha.122.036946] [Citation(s) in RCA: 303] [Impact Index Per Article: 101.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Maintaining blood-brain barrier (BBB) integrity is crucial for the homeostasis of the central nervous system. Structurally comprising the BBB, brain endothelial cells interact with pericytes, astrocytes, neurons, microglia, and perivascular macrophages in the neurovascular unit. Brain ischemia unleashes a profound neuroinflammatory response to remove the damaged tissue and prepare the brain for repair. However, the intense neuroinflammation occurring during the acute phase of stroke is associated with BBB breakdown, neuronal injury, and worse neurological outcomes. Here, we critically discuss the role of neuroinflammation in ischemic stroke pathology, focusing on the BBB and the interactions between central nervous system and peripheral immune responses. We highlight inflammation-driven injury mechanisms in stroke, including oxidative stress, increased MMP (matrix metalloproteinase) production, microglial activation, and infiltration of peripheral immune cells into the ischemic tissue. We provide an updated overview of imaging techniques for in vivo detection of BBB permeability, leukocyte infiltration, microglial activation, and upregulation of cell adhesion molecules following ischemic brain injury. We discuss the possibility of clinical implementation of imaging modalities to assess stroke-associated neuroinflammation with the potential to provide image-guided diagnosis and treatment. We summarize the results from several clinical studies evaluating the efficacy of anti-inflammatory interventions in stroke. Although convincing preclinical evidence suggests that neuroinflammation is a promising target for ischemic stroke, thus far, translating these results into the clinical setting has proved difficult. Due to the dual role of inflammation in the progression of ischemic damage, more research is needed to mechanistically understand when the neuroinflammatory response begins the transition from injury to repair. This could have important implications for ischemic stroke treatment by informing time- and context-specific therapeutic interventions.
Collapse
Affiliation(s)
- Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville (E.C-J)
| | - Rick M Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht, the Netherlands (R.M.D.)
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Germany (T.M.)
| |
Collapse
|
14
|
Schädlich IS, Vienhues JH, Jander A, Piepke M, Magnus T, Lambertsen KL, Clausen BH, Gelderblom M. Interleukin-1 Mediates Ischemic Brain Injury via Induction of IL-17A in γδ T Cells and CXCL1 in Astrocytes. Neuromolecular Med 2022; 24:437-451. [PMID: 35384588 PMCID: PMC9684245 DOI: 10.1007/s12017-022-08709-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/04/2022] [Indexed: 11/29/2022]
Abstract
As a prototypical proinflammatory cytokine, interleukin-1 (IL-1) exacerbates the early post-stroke inflammation, whereas its neutralization is protective. To further investigate the underlying cell-type-specific IL-1 effects, we subjected IL-1 (α/β) knockout (Il1−/−) and wildtype (WT) littermate mice to permanent middle cerebral artery occlusion (pMCAO) and assessed immune cell infiltration and cytokine production in the ischemic hemisphere by flow cytometry 24 h and 72 h after stroke. Il1−/− mice showed smaller infarcts and reduced neutrophil infiltration into the ischemic brain. We identified γδ T cells and astrocytes as target cells of IL-1 signaling-mediated neutrophil recruitment. First, IL-1-induced IL-17A production in γδ T cells in vivo, and IL-17A enhanced the expression of the main neutrophil attracting chemokine CXCL1 by astrocytes in the presence of tumor necrosis factor (TNF) in vitro. Second, IL-1 itself was a potent activator of astrocytic CXCL1 production in vitro. By employing a novel FACS sorting strategy for the acute isolation of astrocytes from ischemic brains, we confirmed that IL-1 is pivotal for Cxcl1 upregulation in astrocytes in vivo. Our results underscore the pleiotropic effects of IL-1 on immune and non-immune cells within the CNS to mount and amplify the post-stroke inflammatory response.
Collapse
Affiliation(s)
- Ines Sophie Schädlich
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg-Eppendorf, Germany.
| | - Jonas Heinrich Vienhues
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg-Eppendorf, Germany.,Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Alina Jander
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg-Eppendorf, Germany
| | - Marius Piepke
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg-Eppendorf, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg-Eppendorf, Germany
| | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE - Brain Research - Inter Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Deparment of Neurology, Odense University Hospital, Odense, Denmark
| | - Bettina Hjelm Clausen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE - Brain Research - Inter Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg-Eppendorf, Germany
| |
Collapse
|
15
|
Russell AAM, Sutherland BA, Landowski LM, Macleod M, Howells DW. What has preclinical systematic review ever done for us? BMJ OPEN SCIENCE 2022; 6:e100219. [PMID: 35360370 PMCID: PMC8921935 DOI: 10.1136/bmjos-2021-100219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
Systematic review and meta-analysis are a gift to the modern researcher, delivering a crystallised understanding of the existing research data in any given space. This can include whether candidate drugs are likely to work or not and which are better than others, whether our models of disease have predictive value and how this might be improved and also how these all interact with disease pathophysiology.Grappling with the literature needed for such analyses is becoming increasingly difficult as the number of publications grows. However, narrowing the focus of a review to reduce workload runs the risk of diminishing the generalisability of conclusions drawn from such increasingly specific analyses.Moreover, at the same time as we gain greater insight into our topic, we also discover more about the flaws that undermine much scientific research. Systematic review and meta-analysis have also shown that the quality of much preclinical research is inadequate. Systematic review has helped reveal the extent of selection bias, performance bias, detection bias, attrition bias and low statistical power, raising questions about the validity of many preclinical research studies. This is perhaps the greatest virtue of systematic review and meta-analysis, the knowledge generated ultimately helps shed light on the limitations of existing research practice, and in doing so, helps bring reform and rigour to research across the sciences.In this commentary, we explore the lessons that we have identified through the lens of preclinical systematic review and meta-analysis.
Collapse
Affiliation(s)
- Ash Allanna Mark Russell
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Brad A Sutherland
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Lila M Landowski
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Malcolm Macleod
- Centre for Clinical Brain Sciences, Edinburgh Medical School, The University of Edinburgh, Edinburgh, UK
| | - David W Howells
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
16
|
Tanriver-Ayder E, Faes C, van de Casteele T, McCann SK, Macleod MR. Comparison of commonly used methods in random effects meta-analysis: application to preclinical data in drug discovery research. BMJ OPEN SCIENCE 2022; 5:e100074. [PMID: 35047696 PMCID: PMC8647574 DOI: 10.1136/bmjos-2020-100074] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 11/26/2020] [Accepted: 01/08/2021] [Indexed: 11/25/2022] Open
Abstract
Background Meta-analysis of preclinical data is used to evaluate the consistency of findings and to inform the design and conduct of future studies. Unlike clinical meta-analysis, preclinical data often involve many heterogeneous studies reporting outcomes from a small number of animals. Here, we review the methodological challenges in preclinical meta-analysis in estimating and explaining heterogeneity in treatment effects. Methods Assuming aggregate-level data, we focus on two topics: (1) estimation of heterogeneity using commonly used methods in preclinical meta-analysis: method of moments (DerSimonian and Laird; DL), maximum likelihood (restricted maximum likelihood; REML) and Bayesian approach; (2) comparison of univariate versus multivariable meta-regression for adjusting estimated treatment effects for heterogeneity. Using data from a systematic review on the efficacy of interleukin-1 receptor antagonist in animals with stroke, we compare these methods, and explore the impact of multiple covariates on the treatment effects. Results We observed that the three methods for estimating heterogeneity yielded similar estimates for the overall effect, but different estimates for between-study variability. The proportion of heterogeneity explained by a covariate is estimated larger using REML and the Bayesian method as compared with DL. Multivariable meta-regression explains more heterogeneity than univariate meta-regression. Conclusions Our findings highlight the importance of careful selection of the estimation method and the use of multivariable meta-regression to explain heterogeneity. There was no difference between REML and the Bayesian method and both methods are recommended over DL. Multiple meta-regression is worthwhile to explain heterogeneity by more than one variable, reducing more variability than any univariate models and increasing the explained proportion of heterogeneity.
Collapse
Affiliation(s)
- Ezgi Tanriver-Ayder
- Centre for Clinical Brain Sciences, Edinburgh Medical School, The University of Edinburgh, Edinburgh, Scotland, UK.,Translational Medicine and Early Development Statistics, Janssen Pharmaceutica, Beerse, Antwerpen, Belgium
| | - Christel Faes
- Data Science Institute (DSI), Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-BioStat), Hasselt University, Hasselt, Limburg, Belgium
| | - Tom van de Casteele
- Translational Medicine and Early Development Statistics, Janssen Pharmaceutica, Beerse, Antwerpen, Belgium
| | - Sarah K McCann
- QUEST Center for Transforming Biomedical Research, Berlin Institute of Health (BIH), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Malcolm R Macleod
- Centre for Clinical Brain Sciences, Edinburgh Medical School, The University of Edinburgh, Edinburgh, Scotland, UK
| |
Collapse
|
17
|
Effect of transcranial magnetic stimulation on treatment effect and immune function. Saudi J Biol Sci 2022; 29:379-384. [PMID: 35002433 PMCID: PMC8717157 DOI: 10.1016/j.sjbs.2021.08.104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/29/2021] [Accepted: 08/31/2021] [Indexed: 11/20/2022] Open
Abstract
To explore the effect of transcranial stimulation on the therapeutic effect and immune function of patients with post-stroke depression (PSD). Methods Selection in September 2020-April 2021 on the diagnosis of 70 patients with PSD as the research object, 35 patients were randomly divided into control group and intervention group and control group given conventional treatment, the intervention group in the control group on the basis of the application of transcranial magnetic stimulation treatment, compare the curative effect of two groups of patients after the treatment cycle and the effects on the immune function. Results After treatment, the levels of DA, NE, 5-HT in 2 groups were significantly increased, and those in the observation group were significantly higher than those in the control group (P < 0.05). After 8 weeks of treatment, serum Gly content in 2 groups was significantly increased and Glu content was significantly decreased compared with before treatment. Compared with the control group, serum Gly content in observation group was significantly increased and Glu content was significantly decreased after treatment (P < 0.05). After 8 weeks of treatment, the contents of IL-1β, IL-6 and TNF-α in serum of 2 groups were significantly decreased, compared with the control group, the contents of IL-1β, IL-6 and TNF-α in serum of observation group were significantly decreased (P < 0.05); Before treatment, there was no significant difference in PHQ-9 score and MBI score between the two groups (P > 0.05). After 8 weeks of treatment, PHQ-9 score and MBI score in the two groups were better than before treatment, and the observation group was better than the control group (P < 0.05). Conclusion Transcranial magnetic stimulation therapy can not only effectively promote the synthesis and release of monoamine neurotransmitters in patients with post-stroke depression, regulate the inhibitory/excitatory amino acid neurotransmitters, reduce inflammatory response, improve the clinical treatment effect and enhance the immune function of PSD patients, which has clinical application value.
Collapse
|
18
|
Stuckey SM, Ong LK, Collins-Praino LE, Turner RJ. Neuroinflammation as a Key Driver of Secondary Neurodegeneration Following Stroke? Int J Mol Sci 2021; 22:ijms222313101. [PMID: 34884906 PMCID: PMC8658328 DOI: 10.3390/ijms222313101] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/25/2021] [Accepted: 12/01/2021] [Indexed: 01/13/2023] Open
Abstract
Ischaemic stroke involves the rapid onset of focal neurological dysfunction, most commonly due to an arterial blockage in a specific region of the brain. Stroke is a leading cause of death and common cause of disability, with over 17 million people worldwide suffering from a stroke each year. It is now well-documented that neuroinflammation and immune mediators play a key role in acute and long-term neuronal tissue damage and healing, not only in the infarct core but also in distal regions. Importantly, in these distal regions, termed sites of secondary neurodegeneration (SND), spikes in neuroinflammation may be seen sometime after the initial stroke onset, but prior to the presence of the neuronal tissue damage within these regions. However, it is key to acknowledge that, despite the mounting information describing neuroinflammation following ischaemic stroke, the exact mechanisms whereby inflammatory cells and their mediators drive stroke-induced neuroinflammation are still not fully understood. As a result, current anti-inflammatory treatments have failed to show efficacy in clinical trials. In this review we discuss the complexities of post-stroke neuroinflammation, specifically how it affects neuronal tissue and post-stroke outcome acutely, chronically, and in sites of SND. We then discuss current and previously assessed anti-inflammatory therapies, with a particular focus on how failed anti-inflammatories may be repurposed to target SND-associated neuroinflammation.
Collapse
Affiliation(s)
- Shannon M. Stuckey
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
| | - Lin Kooi Ong
- School of Pharmacy, Monash University Malaysia, Subang Jaya 47500, Malaysia;
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, The University of Newcastle, Callaghan 2308, Australia
| | - Lyndsey E. Collins-Praino
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
| | - Renée J. Turner
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
- Correspondence: ; Tel.: +61-8-8313-3114
| |
Collapse
|
19
|
Sjöström EO, Culot M, Leickt L, Åstrand M, Nordling E, Gosselet F, Kaiser C. Transport study of interleukin-1 inhibitors using a human in vitro model of the blood-brain barrier. Brain Behav Immun Health 2021; 16:100307. [PMID: 34589799 PMCID: PMC8474601 DOI: 10.1016/j.bbih.2021.100307] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/24/2021] [Indexed: 01/15/2023] Open
Abstract
The proinflammatory cytokine Interleukin-1 (IL-1), with its two isoforms α and β, has important roles in multiple pathogenic processes in the central nervous system. The present study aimed to evaluate and compare the blood-to-brain distribution of anakinra (IL-1 receptor antagonist), bermekimab (IL-1α antagonist) and canakinumab (IL-1β antagonist). A human in vitro model of the blood-brain barrier derived from human umbilical cord blood stem cells was used, where isolated CD34+ cells co-cultured with bovine pericytes were matured into polarized brain-like endothelial cells. Transport rates of the three test items were evaluated after 180 min incubation at concentrations 50, 250 and 1250 nM in a transwell system. We report herein that anakinra passes the human brain-like endothelial monolayer at a 4-7-fold higher rate than the monoclonal antibodies tested. Both antibodies had similar transport rates at all concentrations. No dose-dependent effects in transport rates were observed, nor any saturation effects at supraphysiological concentrations. The larger propensity of anakinra to pass this model of the human blood-brain barrier supports existing data and confirms that anakinra can reach the brain compartment at clinically relevant concentrations. As anakinra inhibits the actions of both IL-1α and IL-1β, it blocks all effects of IL-1 downstream signaling. The results herein further add to the growing body of evidence of the potential utility of anakinra to treat neuroinflammatory disorders. Anakinra has a larger propensity to pass the in vitro BBB than monoclonal antibodies targeting the IL-1 system. Implications for targeting inflammation in cerebral ischemia and neurological sequelae of autoinflammatory diseases. Novel and comparative study of biologics in a human in vitro BBB model shows relevance and validity.
Collapse
Affiliation(s)
| | - Maxime Culot
- Univ. Artois, UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), F-62300, Lens, France
| | - Lisa Leickt
- Swedish Orphan Biovitrum AB (publ), SE-112 76, Stockholm, Sweden
| | - Mikael Åstrand
- Swedish Orphan Biovitrum AB (publ), SE-112 76, Stockholm, Sweden
| | - Erik Nordling
- Swedish Orphan Biovitrum AB (publ), SE-112 76, Stockholm, Sweden
| | - Fabien Gosselet
- Univ. Artois, UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), F-62300, Lens, France
| | - Christina Kaiser
- Swedish Orphan Biovitrum AB (publ), SE-112 76, Stockholm, Sweden
| |
Collapse
|
20
|
Venugopal J, Wang J, Mawri J, Guo C, Eitzman D. Interleukin-1 receptor inhibition reduces stroke size in a murine model of sickle cell disease. Haematologica 2021; 106:2469-2477. [PMID: 32817286 PMCID: PMC8409048 DOI: 10.3324/haematol.2020.252395] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Indexed: 12/22/2022] Open
Abstract
Sickle cell disease (SCD) is associated with chronic hemolytic anemia and a heightened inflammatory state. The causal role of inflammatory pathways in stroke associated with SCD is unclear. Therefore, the hypothesis that deletion of the non-hematopoietic interleukin-1 receptor (IL-1R) pool may be beneficial in SCD was pursued. Since potential deleterious effects of IL-1R signaling in SCD could be mediated via downstream production of interleukin-6 (IL-6), the role of the nonhematopoietic IL-6 pool was also addressed. Bone marrow transplantation (BMT) from SCD to wild-type (WT) recipient mice was used to generate SCD mice (Wt,SCDbmt). In order to generate mice with nonhematopoietic deficiency of IL-1R or IL-6, SCD marrow was transplanted into IL-1R deficient (IL1R-/-,SCDbmt) or IL-6 deficient recipients (IL6-/-, SCDbmt). Blood counts, reticulocytes, soluble E-selectin (sEsel), and IL-6 levels were analyzed 14-15 weeks post-BMT. Ischemic stroke was induced by middle cerebral artery (MCA) photothrombosis at 16 weeks post-BMT. A separate group of Wt,SCDbmt mice was given the IL-1R inhibitor, anakinra, following stroke induction. Seventy-two hours after MCA occlusion, stroke volume was assessed by staining brain sections with 2,3,5-triphenyltetrazolium chloride. Formalin-fixed brain sections were also stained for macrophages with MAC3, for endothelial activation with ICAM-1, and for loss of blood brain barrier integrity with fibrin (ogen) staining. All SCD mice generated by BMT were anemic and the severity of anemia was not different between Wt,SCDbmt, IL1R-/-,SCDbmt, and IL-6-/-,SCDbmt mice. Three days following MCA occlusion, stroke volume was significantly reduced in IL1R-/-,SCDbmt mice compared to Wt,SCDbmt mice and IL6-/-,SCDbmt mice. Plasma levels of sEsel were lower in IL1R-/-,SCDbmt compared to Wt,SCDbmt and IL-6-/-,SCDbmt mice. Post-stroke treatment of Wt,SCDbmt mice with anakinra decreased stroke size, leukocyte infiltration, ICAM-1 expression, and fibrin(ogen) accumulation compared to vehicle-treated mice. Deficiency of non-hematopoietic IL-1R or treatment with an IL-1R antagonist is sufficient to confer protection against the increased stroke size associated with SCD. These effects of IL1R deficiency are associated with reduced endothelial activation, leukocyte infiltration, and blood brain barrier disruption, and are independent of non-hematopoietic IL-6 signaling.
Collapse
Affiliation(s)
- Jessica Venugopal
- University of Michigan Internal Medicine - Cardiology division, Ann Arbor, MI, USA
| | - Jintao Wang
- University of Michigan Internal Medicine - Cardiology division, Ann Arbor, MI, USA
| | | | - Chiao Guo
- University of Michigan Internal Medicine - Cardiology division, Ann Arbor, MI, USA
| | - Daniel Eitzman
- University of Michigan Internal Medicine - Cardiology division, Ann Arbor, MI, USA
| |
Collapse
|
21
|
Abstract
We search for ischemic stroke treatment knowing we have failed-intensely and often-to translate mechanistic knowledge into treatments that alleviate our patients' functional impairments. Lessons can be derived from our shared failures that may point to new directions and new strategies. First, the principle criticisms of both preclinical and clinical assessments are summarized. Next, previous efforts to develop single-mechanism treatments are reviewed. Finally, new definitions, novel approaches, and different directions are presented. In previous development efforts, the basic science and preclinical assessment of candidate treatments often lacked rigor and sufficiency; the clinical trials may have lacked power, rigor, or rectitude; or most likely both preclinical and clinical investigations were flawed. Single-target agents directed against specific molecular mechanisms proved unsuccessful. The term neuroprotection should be replaced as it has become ambiguous: protection of the entire neurovascular unit may be called cerebral cytoprotection or cerebroprotection. Success in developing cerebroprotection-either as an adjunct to recanalization or as stand-alone treatment-will require new definitions that recognize the importance of differential vulnerability in the neurovascular unit. Recent focus on pleiotropic multi-target agents that act via multiple mechanisms of action to interrupt ischemia at multiple steps may be more fruitful. Examples of pleiotropic treatments include therapeutic hypothermia and 3K3A-APC (activated protein C). Alternatively, the single-target drug NA-1 triggers multiple downstream signaling events. Renewed commitment to scientific rigor is essential, and funding agencies and journals may enforce quality principles of rigor in preclinical science. Appropriate animal models should be selected that are suited to the purpose of the investigation. Before clinical trials, preclinical assessment could include subjects that are aged, of both sexes, and harbor comorbid conditions such as diabetes or hypertension. With these new definitions, novel approaches, and renewed attention to rigor, the prospect for successful cerebroprotective therapy should improve.
Collapse
Affiliation(s)
- Patrick D Lyden
- Department of Physiology and Neuroscience, Department of Neurology, Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, CA
| |
Collapse
|
22
|
Myers AK, Talbot CF, Del Rosso LA, Maness AC, Simmons SMV, Garner JP, Capitanio JP, Parker KJ. Assessment of medical morbidities in a rhesus monkey model of naturally occurring low sociality. Autism Res 2021; 14:1332-1346. [PMID: 33847078 DOI: 10.1002/aur.2512] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/27/2021] [Accepted: 03/22/2021] [Indexed: 12/27/2022]
Abstract
People with autism spectrum disorder (ASD) exhibit a variety of medical morbidities at significantly higher rates than the general population. Using an established monkey model of naturally occurring low sociality, we investigated whether low-social monkeys show an increased burden of medical morbidities compared to their high-social counterparts. We systematically reviewed the medical records of N = 152 (n = 73 low-social; n = 79 high-social) rhesus macaques (Macaca mulatta) to assess the number of traumatic injury, gastrointestinal, and inflammatory events, as well as the presence of rare medical conditions. Subjects' nonsocial scores, determined by the frequency they were observed in a nonsocial state (i.e., alone), and macaque Social Responsiveness Scale-Revised (mSRS-R) scores were also used to test whether individual differences in social functioning were related to medical morbidity burden. Medical morbidity type significantly differed by group, such that low-social monkeys incurred higher rates of traumatic injury compared to high-social monkeys. Nonsocial scores and mSRS-R scores also significantly and positively predicted traumatic injury rates, indicating that monkeys with the greatest social impairment were most impacted on this health measure. These findings from low-social monkeys are consistent with well-documented evidence that people with ASD incur a greater number of traumatic injuries and receive more peer bullying than their neurotypical peers, and add to growing evidence for the face validity of this primate model. LAY SUMMARY: People with autism exhibit multiple medical problems at higher rates than the general population. We conducted a comprehensive medical record review of monkeys that naturally exhibit differences in sociality and found that low-social monkeys are more susceptible to traumatic injuries than high-social monkeys. These results are consistent with reports that people with autism also incur greater traumatic injury and peer bullying and add to growing evidence for the validity of this monkey model.
Collapse
Affiliation(s)
- Adam K Myers
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California, USA.,College of Veterinary Medicine, Western University of Health Sciences, Pomona, California, USA
| | - Catherine F Talbot
- California National Primate Research Center, University of California, Davis, California, USA
| | - Laura A Del Rosso
- California National Primate Research Center, University of California, Davis, California, USA
| | - Alyssa C Maness
- California National Primate Research Center, University of California, Davis, California, USA
| | - Sierra M V Simmons
- California National Primate Research Center, University of California, Davis, California, USA
| | - Joseph P Garner
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California, USA.,Department of Comparative Medicine, Stanford University, Stanford, California, USA
| | - John P Capitanio
- California National Primate Research Center, University of California, Davis, California, USA.,Department of Psychology, University of California, Davis, California, USA
| | - Karen J Parker
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California, USA.,California National Primate Research Center, University of California, Davis, California, USA
| |
Collapse
|
23
|
Aliena-Valero A, Baixauli-Martín J, Castelló-Ruiz M, Torregrosa G, Hervás D, Salom JB. Effect of uric acid in animal models of ischemic stroke: A systematic review and meta-analysis. J Cereb Blood Flow Metab 2021; 41:707-722. [PMID: 33210575 PMCID: PMC7983496 DOI: 10.1177/0271678x20967459] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Addition of uric acid (UA) to thrombolytic therapy, although safe, showed limited efficacy in improving patients' stroke outcome, despite alleged neuroprotective effects of UA in preclinical research. This systematic review assessed the effects of UA on brain structural and functional outcomes in animal models of ischemic stroke. We searched Medline, Embase and Web of Science to identify 16 and 14 eligible rodent studies for qualitative and quantitative synthesis, respectively. Range of evidence met 10 of a possible 13 STAIR criteria. Median (Q1, Q3) quality score was 7.5 (6, 10) on the CAMARADES 15-item checklist. For each outcome, we used standardised mean difference (SMD) as effect size and random-effects modelling. Meta-analysis showed that UA significantly reduced infarct size (SMD: -1.18; 95% CI [-1.47, -0.88]; p < 0.001), blood-brain barrier (BBB) impairment/oedema (SMD: -0.72; 95% CI [-0.97, -0.48]; p < 0.001) and neurofunctional deficit (SMD: -0.98; 95% CI [-1.32, -0.63]; p < 0.001). Overall, there was low to moderate between-study heterogeneity and sizeable publication bias. In conclusion, published rodent data suggest that UA improves outcome following ischemic stroke by reducing infarct size, improving BBB integrity and ameliorating neurofunctional condition. Specific recommendations are given for further high-quality preclinical research required to better inform clinical research.
Collapse
Affiliation(s)
- Alicia Aliena-Valero
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universidad de Valencia, Valencia, Spain
| | | | - María Castelló-Ruiz
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universidad de Valencia, Valencia, Spain.,Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, Valencia, Spain
| | - Germán Torregrosa
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universidad de Valencia, Valencia, Spain
| | - David Hervás
- Unidad de Bioestadística, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Juan B Salom
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universidad de Valencia, Valencia, Spain.,Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| |
Collapse
|
24
|
Bahor Z, Liao J, Currie G, Ayder C, Macleod M, McCann SK, Bannach-Brown A, Wever K, Soliman N, Wang Q, Doran-Constant L, Young L, Sena ES, Sena C. Development and uptake of an online systematic review platform: the early years of the CAMARADES Systematic Review Facility (SyRF). BMJ OPEN SCIENCE 2021; 5:e100103. [PMID: 35047698 PMCID: PMC8647599 DOI: 10.1136/bmjos-2020-100103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 01/20/2023] Open
Abstract
Preclinical research is a vital step in the drug discovery pipeline and more generally in helping to better understand human disease aetiology and its management. Systematic reviews (SRs) can be powerful in summarising and appraising this evidence concerning a specific research question, to highlight areas of improvements, areas for further research and areas where evidence may be sufficient to take forward to other research domains, for instance clinical trial. Guidance and tools for preclinical research synthesis remain limited despite their clear utility. We aimed to create an online end-to-end platform primarily for conducting SRs of preclinical studies, that was flexible enough to support a wide variety of experimental designs, was adaptable to different research questions, would allow users to adopt emerging automated tools and support them during their review process using best practice. In this article, we introduce the Systematic Review Facility (https://syrf.org.uk), which was launched in 2016 and designed to support primarily preclinical SRs from small independent projects to large, crowdsourced projects. We discuss the architecture of the app and its features, including the opportunity to collaborate easily, to efficiently manage projects, to screen and annotate studies for important features (metadata), to extract outcome data into a secure database, and tailor these steps to each project. We introduce how we are working to leverage the use of automation tools and allow the integration of these services to accelerate and automate steps in the systematic review workflow.
Collapse
Affiliation(s)
- Zsanett Bahor
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Jing Liao
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Gillian Currie
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Can Ayder
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Malcolm Macleod
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Sarah K McCann
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- QUEST - Center for Transforming Biomedical Research, Berlin Institute of Health (BIH), Berlin, Germany
| | - Alexandra Bannach-Brown
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland, UK
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- QUEST - Center for Transforming Biomedical Research, Berlin Institute of Health (BIH), Berlin, Germany
- Institute for Evidence-Based Practice, Bond University, Robina, Queensland, Australia
| | - Kimberley Wever
- Systematic Review Centre for Laboratory animal Experimentation (SYRCLE), Department for Health Evidence, Nijmegen Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Qianying Wang
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland, UK
| | | | | | - Emily S Sena
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Chris Sena
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland, UK
| |
Collapse
|
25
|
Cicolari S, Catapano AL, Magni P. Inflammaging and neurodegenerative diseases: Role of NLRP3 inflammasome activation in brain atherosclerotic vascular disease. Mech Ageing Dev 2021; 195:111467. [PMID: 33711349 DOI: 10.1016/j.mad.2021.111467] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 02/26/2021] [Accepted: 03/07/2021] [Indexed: 02/07/2023]
Abstract
The activation of the NLRP3 inflammasome-IL-1β pathway has been clearly shown to be involved in the pathophysiology of cardiovascular diseases, but its role in cerebral atherosclerotic vascular disease has not been fully clarified. Here we provide an overview on the current knowledge about the relevance of the activation of this mechanism in the onset of acute brain atherosclerotic vascular disease and the subsequent tissue damage. Some variants of NLRP3-related genes seem to reduce the susceptibility to acute ischaemic stroke in selected cohorts, although no clear evidence exists either supporting or excluding any role of this pathway in its pathophysiology. Interestingly, robust experimental and clinical data support a major role of the activation of the NLRP3 inflammasome-IL-1β pathway in the post-event inflammatory cascade which leads to neurodegeneration. This evidence highlights a potential dual role of these molecules in brain pre- and post-ischaemic events, supporting the need for further studies, including clinical trials evaluating the modulation of this pathway for stroke prevention and post-stroke treatment.
Collapse
Affiliation(s)
- Stefania Cicolari
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133, Milan, Italy
| | - Alberico L Catapano
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133, Milan, Italy; IRCCS Multimedica Hospital, 20099, Milan, Italy
| | - Paolo Magni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133, Milan, Italy; IRCCS Multimedica Hospital, 20099, Milan, Italy.
| |
Collapse
|
26
|
Kannan A, Delgardo M, Pennington-FitzGerald W, Jiang EX, Christophe BR, Connolly ES. Pharmacological management of cerebral ischemia in the elderly. Expert Opin Pharmacother 2020; 22:897-906. [PMID: 33382005 DOI: 10.1080/14656566.2020.1856815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: For elderly adults in the United States, stroke is the fifth leading cause of death of which ischemic strokes comprise a vast majority. Optimal pharmacological management of elderly ischemic stroke patients involves both reperfusion and supportive care. Recent research into pharmacological management has focused on vascular, immunomodulatory, cytoprotective, and alternative agents, some of which have shown limited success in clinical trials. However, no treatments have been established as a reliable mode for management of cerebral ischemia for elderly adults beyond acute thrombolysis.Areas covered: The authors conducted a literature search for ischemic stroke management in the elderly and a search for human drug studies for managing ischemic stroke on clinicaltrials.gov. Here, they describe recent progress in the pharmacological management of cerebral ischemia in the elderly.Expert opinion: Many drug classes (antihypertensive, cytoprotective and immunomodulatory, and alternative agents) have been explored with limited success in managing ischemic stroke, though some have shown preventative benefits. We generally observed a broad gap in evidence on elderly patients from studies across all drug classes, necessitating further studies to gain an understanding of effective management of ischemic stroke in this large demographic of patients.
Collapse
Affiliation(s)
- Adithya Kannan
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| | - Mychael Delgardo
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| | | | - Enoch X Jiang
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| | - Brandon R Christophe
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| | - E Sander Connolly
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
27
|
Woods D, Jiang Q, Chu XP. Monoclonal antibody as an emerging therapy for acute ischemic stroke. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2020; 12:95-106. [PMID: 32934765 PMCID: PMC7486556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 08/20/2020] [Indexed: 06/11/2023]
Abstract
Acute ischemic stroke (AIS) is the 5th leading cause of death and the leading cause of neurological disability in the United States. The oxygen and glucose deprivation associated with AIS not only leads to neuronal cell death, but also increases the inflammatory response, therefore decreasing the functional outcome of the brain. The only pharmacological intervention approved by the US Federal Food and Drug Administration for treatment of AIS is tissue plasminogen activator (t-PA), however, such treatment can only be given within 4.5 hours of the onset of stroke-like symptoms. This narrow time-range limits its therapeutic application. Administrating t-PA outside of the therapeutic window may induce detrimental rather than beneficial effects to stroke patients. In order to reduce the infarct volume of an AIS while increasing the time period for treatment, new treatments are essential. Emerging monoclonal antibody (mAb) therapies reveal great potential by targeting signaling pathways activated after an AIS. With successful application of mAb in the treatment of cancer, other therapeutic uses for mAb are currently being evaluated. In this review, we will focus on recent advances on AIS therapy by using mAb that targets the signaling cascades and endogenous molecules such as inflammation, growth factors, acid-sensing ion channels, and N-methyl-D-aspartate receptors. Therefore, developing specific mAb to target the signaling pathways of ischemic brain injury will benefit patients being treated for an AIS.
Collapse
Affiliation(s)
- Demi Woods
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City Kansas City, MO 64108, USA
| | - Qian Jiang
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City Kansas City, MO 64108, USA
| | - Xiang-Ping Chu
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City Kansas City, MO 64108, USA
| |
Collapse
|
28
|
O'Reilly ML, Tom VJ. Neuroimmune System as a Driving Force for Plasticity Following CNS Injury. Front Cell Neurosci 2020; 14:187. [PMID: 32792908 PMCID: PMC7390932 DOI: 10.3389/fncel.2020.00187] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/29/2020] [Indexed: 12/15/2022] Open
Abstract
Following an injury to the central nervous system (CNS), spontaneous plasticity is observed throughout the neuraxis and affects multiple key circuits. Much of this spontaneous plasticity can elicit beneficial and deleterious functional outcomes, depending on the context of plasticity and circuit affected. Injury-induced activation of the neuroimmune system has been proposed to be a major factor in driving this plasticity, as neuroimmune and inflammatory factors have been shown to influence cellular, synaptic, structural, and anatomical plasticity. Here, we will review the mechanisms through which the neuroimmune system mediates plasticity after CNS injury. Understanding the role of specific neuroimmune factors in driving adaptive and maladaptive plasticity may offer valuable therapeutic insight into how to promote adaptive plasticity and/or diminish maladaptive plasticity, respectively.
Collapse
Affiliation(s)
- Micaela L O'Reilly
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Veronica J Tom
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
29
|
Clausen BH, Wirenfeldt M, Høgedal SS, Frich LH, Nielsen HH, Schrøder HD, Østergaard K, Finsen B, Kristensen BW, Lambertsen KL. Characterization of the TNF and IL-1 systems in human brain and blood after ischemic stroke. Acta Neuropathol Commun 2020; 8:81. [PMID: 32503645 PMCID: PMC7273684 DOI: 10.1186/s40478-020-00957-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/27/2020] [Indexed: 01/17/2023] Open
Abstract
Preclinical and clinical proof-of-concept studies have suggested the effectiveness of pharmacological modulation of inflammatory cytokines in ischemic stroke. Experimental evidence shows that targeting tumor necrosis factor (TNF) and interleukin (IL)-1 holds promise, and these cytokines are considered prime targets in the development of new stroke therapies. So far, however, information on the cellular expression of TNF and IL-1 in the human ischemic brain is sparse.We studied 14 cases of human post-mortem ischemic stroke, representing 21 specimens of infarcts aged 1 to > 8 days. We characterized glial and leukocyte reactions in the infarct/peri-infarct (I/PI) and normal-appearing tissue (NAT) and the cellular location of TNF, TNF receptor (TNFR)1 and TNFR2, IL-1α, IL-1β, and IL-1 receptor antagonist (IL-1Ra). The immunohistochemically stained tissue sections received a score reflecting the number of immunoreactive cells and the intensity of the immunoreactivity (IR) in individual cells where 0 = no immunoreactive cells, 1 = many intermediately to strongly immunoreactive cells, and 2 = numerous and intensively immunoreactive cells. Additionally, we measured blood TNF, TNFR, and IL-1 levels in surviving ischemic stroke patients within the first 8 h and again at 72 h after symptom onset and compared levels to healthy controls.We observed IL-1α and IL-1β IR in neurons, glia, and macrophages in all specimens. IL-1Ra IR was found in glia, in addition to macrophages. TNF IR was initially found in neurons located in I/PI and NAT but increased in glia in older infarcts. TNF IR increased in macrophages in all specimens. TNFR1 IR was found in neurons and glia and macrophages, while TNFR2 was expressed only by glia in I/PI and NAT, and by macrophages in I/PI. Our results suggest that TNF and IL-1 are expressed by subsets of cells and that TNFR2 is expressed in areas with increased astrocytic reactivity. In ischemic stroke patients, we demonstrate that plasma TNFR1 and TNFR2 levels increased in the acute phase after symptom onset compared to healthy controls, whereas TNF, IL-1α, IL-1β, and IL-1Ra did not change.Our findings of increased brain cytokines and plasma TNFR1 and TNFR2 support the hypothesis that targeting post-stroke inflammation could be a promising add-on therapy in ischemic stroke patients.
Collapse
Affiliation(s)
- Bettina H. Clausen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21, st, DK-5000 Odense C, Denmark
- BRIDGE, Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark
| | - Martin Wirenfeldt
- BRIDGE, Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark
- Department of Pathology, Odense University Hospital, Odense, J.B. Winsloewsvej 15, DK-5000 Odense C, Denmark
| | - Sofie S. Høgedal
- Department of Neurology, Odense University Hospital, J.B. Winsloewsvej 4, DK-5000 Odense C, Denmark
| | - Lars H. Frich
- Orthopedic Research Unit, University of Southern Denmark, DK-5000 Odense C, Denmark
- OPEN, Open Patient data Explorative Network, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, J.B. Winsloewsvej 9a, DK-5000 Odense, Denmark
| | - Helle H. Nielsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21, st, DK-5000 Odense C, Denmark
- BRIDGE, Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark
- Department of Neurology, Odense University Hospital, J.B. Winsloewsvej 4, DK-5000 Odense C, Denmark
| | - Henrik D. Schrøder
- Department of Pathology, Odense University Hospital, Odense, J.B. Winsloewsvej 15, DK-5000 Odense C, Denmark
| | - Kamilla Østergaard
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21, st, DK-5000 Odense C, Denmark
| | - Bente Finsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21, st, DK-5000 Odense C, Denmark
- BRIDGE, Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark
| | - Bjarne W. Kristensen
- BRIDGE, Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark
- Department of Pathology, Odense University Hospital, Odense, J.B. Winsloewsvej 15, DK-5000 Odense C, Denmark
| | - Kate L. Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21, st, DK-5000 Odense C, Denmark
- BRIDGE, Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark
- Department of Neurology, Odense University Hospital, J.B. Winsloewsvej 4, DK-5000 Odense C, Denmark
- OPEN, Open Patient data Explorative Network, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, J.B. Winsloewsvej 9a, DK-5000 Odense, Denmark
| |
Collapse
|
30
|
Applying univariate vs. multivariate statistics to investigate therapeutic efficacy in (pre)clinical trials: A Monte Carlo simulation study on the example of a controlled preclinical neurotrauma trial. PLoS One 2020; 15:e0230798. [PMID: 32214370 PMCID: PMC7098614 DOI: 10.1371/journal.pone.0230798] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 03/09/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Small sample sizes combined with multiple correlated endpoints pose a major challenge in the statistical analysis of preclinical neurotrauma studies. The standard approach of applying univariate tests on individual response variables has the advantage of simplicity of interpretation, but it fails to account for the covariance/correlation in the data. In contrast, multivariate statistical techniques might more adequately capture the multi-dimensional pathophysiological pattern of neurotrauma and therefore provide increased sensitivity to detect treatment effects. RESULTS We systematically evaluated the performance of univariate ANOVA, Welch's ANOVA and linear mixed effects models versus the multivariate techniques, ANOVA on principal component scores and MANOVA tests by manipulating factors such as sample and effect size, normality and homogeneity of variance in computer simulations. Linear mixed effects models demonstrated the highest power when variance between groups was equal or variance ratio was 1:2. In contrast, Welch's ANOVA outperformed the remaining methods with extreme variance heterogeneity. However, power only reached acceptable levels of 80% in the case of large simulated effect sizes and at least 20 measurements per group or moderate effects with at least 40 replicates per group. In addition, we evaluated the capacity of the ordination techniques, principal component analysis (PCA), redundancy analysis (RDA), linear discriminant analysis (LDA), and partial least squares discriminant analysis (PLS-DA) to capture patterns of treatment effects without formal hypothesis testing. While LDA suffered from a high false positive rate due to multicollinearity, PCA, RDA, and PLS-DA were robust and PLS-DA outperformed PCA and RDA in capturing a true treatment effect pattern. CONCLUSIONS Multivariate tests do not provide an appreciable increase in power compared to univariate techniques to detect group differences in preclinical studies. However, PLS-DA seems to be a useful ordination technique to explore treatment effect patterns without formal hypothesis testing.
Collapse
|
31
|
Orsini F, Fumagalli S, Császár E, Tóth K, De Blasio D, Zangari R, Lénárt N, Dénes Á, De Simoni MG. Mannose-Binding Lectin Drives Platelet Inflammatory Phenotype and Vascular Damage After Cerebral Ischemia in Mice via IL (Interleukin)-1α. Arterioscler Thromb Vasc Biol 2019; 38:2678-2690. [PMID: 30354247 PMCID: PMC6221395 DOI: 10.1161/atvbaha.118.311058] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Circulating complement factors are activated by tissue damage and contribute to acute brain injury. The deposition of MBL (mannose-binding lectin), one of the initiators of the lectin complement pathway, on the cerebral endothelium activated by ischemia is a major pathogenic event leading to brain injury. The molecular mechanisms through which MBL influences outcome after ischemia are not understood yet. Approach and Results— Here we show that MBL-deficient (MBL−/−) mice subjected to cerebral ischemia display better flow recovery and less plasma extravasation in the brain than wild-type mice, as assessed by in vivo 2-photon microscopy. This results in reduced vascular dysfunction as shown by the shift from a pro- to an anti-inflammatory vascular phenotype associated with MBL deficiency. We also show that platelets directly bind MBL and that platelets from MBL−/− mice have reduced inflammatory phenotype as indicated by reduced IL-1α (interleukin-1α) content, as early as 6 hours after ischemia. Cultured human brain endothelial cells subjected to oxygen-glucose deprivation and exposed to platelets from MBL−/− mice present less cell death and lower CXCL1 (chemokine [C-X-C motif] ligand 1) release (downstream to IL-1α) than those exposed to wild-type platelets. In turn, MBL deposition on ischemic vessels significantly decreases after ischemia in mice treated with IL-1 receptor antagonist compared with controls, indicating a reciprocal interplay between MBL and IL-1α facilitating endothelial damage. Conclusions— We propose MBL as a hub of pathogenic vascular events. It acts as an early trigger of platelet IL-1α release, which in turn favors MBL deposition on ischemic vessels promoting an endothelial pro-inflammatory phenotype.
Collapse
Affiliation(s)
- Franca Orsini
- From the Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy (F.O., S.F., D.D.B., R.Z., M.-G.D.S.)
| | - Stefano Fumagalli
- From the Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy (F.O., S.F., D.D.B., R.Z., M.-G.D.S.)
| | - Eszter Császár
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary (E.C., K.T., N.L., A.D.)
| | - Krisztina Tóth
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary (E.C., K.T., N.L., A.D.)
| | - Daiana De Blasio
- From the Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy (F.O., S.F., D.D.B., R.Z., M.-G.D.S.)
| | - Rosalia Zangari
- From the Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy (F.O., S.F., D.D.B., R.Z., M.-G.D.S.)
| | - Nikolett Lénárt
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary (E.C., K.T., N.L., A.D.)
| | - Ádám Dénes
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary (E.C., K.T., N.L., A.D.)
| | - Maria-Grazia De Simoni
- From the Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy (F.O., S.F., D.D.B., R.Z., M.-G.D.S.)
| |
Collapse
|
32
|
Malone K, Amu S, Moore AC, Waeber C. Immunomodulatory Therapeutic Strategies in Stroke. Front Pharmacol 2019; 10:630. [PMID: 31281252 PMCID: PMC6595144 DOI: 10.3389/fphar.2019.00630] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/16/2019] [Indexed: 12/14/2022] Open
Abstract
The role of immunity in all stages of stroke is increasingly being recognized, from the pathogenesis of risk factors to tissue repair, leading to the investigation of a range of immunomodulatory therapies. In the acute phase of stroke, proposed therapies include drugs targeting pro-inflammatory cytokines, matrix metalloproteinases, and leukocyte infiltration, with a key objective to reduce initial brain cell toxicity. Systemically, the early stages of stroke are also characterized by stroke-induced immunosuppression, where downregulation of host defences predisposes patients to infection. Therefore, strategies to modulate innate immunity post-stroke have garnered greater attention. A complementary objective is to reduce longer-term sequelae by focusing on adaptive immunity. Following stroke onset, the integrity of the blood–brain barrier is compromised, exposing central nervous system (CNS) antigens to systemic adaptive immune recognition, potentially inducing autoimmunity. Some pre-clinical efforts have been made to tolerize the immune system to CNS antigens pre-stroke. Separately, immune cell populations that exhibit a regulatory phenotype (T- and B- regulatory cells) have been shown to ameliorate post-stroke inflammation and contribute to tissue repair. Cell-based therapies, established in oncology and transplantation, could become a strategy to treat the acute and chronic stages of stroke. Furthermore, a role for the gut microbiota in ischaemic injury has received attention. Finally, the immune system may play a role in remote ischaemic preconditioning-mediated neuroprotection against stroke. The development of stroke therapies involving organs distant to the infarct site, therefore, should not be overlooked. This review will discuss the immune mechanisms of various therapeutic strategies, surveying published data and discussing more theoretical mechanisms of action that have yet to be exploited.
Collapse
Affiliation(s)
- Kyle Malone
- Department of Pharmacology and Therapeutics, School of Pharmacy, University College Cork, Cork, Ireland
| | - Sylvie Amu
- Cancer Research @UCC, University College Cork, Cork, Ireland
| | - Anne C Moore
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Christian Waeber
- Department of Pharmacology and Therapeutics, School of Pharmacy, University College Cork, Cork, Ireland
| |
Collapse
|
33
|
Currie GL, Angel-Scott HN, Colvin L, Cramond F, Hair K, Khandoker L, Liao J, Macleod M, McCann SK, Morland R, Sherratt N, Stewart R, Tanriver-Ayder E, Thomas J, Wang Q, Wodarski R, Xiong R, Rice ASC, Sena ES. Animal models of chemotherapy-induced peripheral neuropathy: A machine-assisted systematic review and meta-analysis. PLoS Biol 2019; 17:e3000243. [PMID: 31107871 PMCID: PMC6544332 DOI: 10.1371/journal.pbio.3000243] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 05/31/2019] [Accepted: 04/08/2019] [Indexed: 12/25/2022] Open
Abstract
We report a systematic review and meta-analysis of research using animal models of chemotherapy-induced peripheral neuropathy (CIPN). We systematically searched 5 online databases in September 2012 and updated the search in November 2015 using machine learning and text mining to reduce the screening for inclusion workload and improve accuracy. For each comparison, we calculated a standardised mean difference (SMD) effect size, and then combined effects in a random-effects meta-analysis. We assessed the impact of study design factors and reporting of measures to reduce risks of bias. We present power analyses for the most frequently reported behavioural tests; 337 publications were included. Most studies (84%) used male animals only. The most frequently reported outcome measure was evoked limb withdrawal in response to mechanical monofilaments. There was modest reporting of measures to reduce risks of bias. The number of animals required to obtain 80% power with a significance level of 0.05 varied substantially across behavioural tests. In this comprehensive summary of the use of animal models of CIPN, we have identified areas in which the value of preclinical CIPN studies might be increased. Using both sexes of animals in the modelling of CIPN, ensuring that outcome measures align with those most relevant in the clinic, and the animal's pain contextualised ethology will likely improve external validity. Measures to reduce risk of bias should be employed to increase the internal validity of studies. Different outcome measures have different statistical power, and this can refine our approaches in the modelling of CIPN.
Collapse
Affiliation(s)
- Gillian L. Currie
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Helena N. Angel-Scott
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Lesley Colvin
- Department of Anaesthesia, Critical Care & Pain, University of Edinburgh, Edinburgh, United Kingdom
- Division of Population Health and Genomics, University of Dundee, Dundee, United Kingdom
| | - Fala Cramond
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Kaitlyn Hair
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Laila Khandoker
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Jing Liao
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Malcolm Macleod
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Sarah K. McCann
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Rosie Morland
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Nicki Sherratt
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Robert Stewart
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Ezgi Tanriver-Ayder
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - James Thomas
- EPPI-Centre, University College London, London, United Kingdom
| | - Qianying Wang
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Rachel Wodarski
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Ran Xiong
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Andrew S. C. Rice
- Pain Research, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Emily S. Sena
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
34
|
Wong R, Lénárt N, Hill L, Toms L, Coutts G, Martinecz B, Császár E, Nyiri G, Papaemmanouil A, Waisman A, Müller W, Schwaninger M, Rothwell N, Francis S, Pinteaux E, Denés A, Allan SM. Interleukin-1 mediates ischaemic brain injury via distinct actions on endothelial cells and cholinergic neurons. Brain Behav Immun 2019; 76:126-138. [PMID: 30453020 PMCID: PMC6363965 DOI: 10.1016/j.bbi.2018.11.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/12/2018] [Accepted: 11/15/2018] [Indexed: 12/17/2022] Open
Abstract
The cytokine interleukin-1 (IL-1) is a key contributor to neuroinflammation and brain injury, yet mechanisms by which IL-1 triggers neuronal injury remain unknown. Here we induced conditional deletion of IL-1R1 in brain endothelial cells, neurons and blood cells to assess site-specific IL-1 actions in a model of cerebral ischaemia in mice. Tamoxifen treatment of IL-1R1 floxed (fl/fl) mice crossed with mice expressing tamoxifen-inducible Cre-recombinase under the Slco1c1 promoter resulted in brain endothelium-specific deletion of IL-1R1 and a significant decrease in infarct size (29%), blood-brain barrier (BBB) breakdown (53%) and neurological deficit (40%) compared to vehicle-treated or control (IL-1R1fl/fl) mice. Absence of brain endothelial IL-1 signalling improved cerebral blood flow, followed by reduced neutrophil infiltration and vascular activation 24 h after brain injury. Conditional IL-1R1 deletion in neurons using tamoxifen inducible nestin-Cre mice resulted in reduced neuronal injury (25%) and altered microglia-neuron interactions, without affecting cerebral perfusion or vascular activation. Deletion of IL-1R1 specifically in cholinergic neurons reduced infarct size, brain oedema and improved functional outcome. Ubiquitous deletion of IL-1R1 had no effect on brain injury, suggesting beneficial compensatory mechanisms on other cells against the detrimental effects of IL-1 on endothelial cells and neurons. We also show that IL-1R1 signalling deletion in platelets or myeloid cells does not contribute to brain injury after experimental stroke. Thus, brain endothelial and neuronal (cholinergic) IL-1R1 mediate detrimental actions of IL-1 in the brain in ischaemic stroke. Cell-specific targeting of IL-1R1 in the brain could therefore have therapeutic benefits in stroke and other cerebrovascular diseases.
Collapse
Affiliation(s)
- Raymond Wong
- Faculty of Biology, Medicine and Health, University of Manchester, M13 9PT Manchester, UK
| | - Nikolett Lénárt
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Szigony u. 43, 1083 Budapest, Hungary
| | - Laura Hill
- Faculty of Biology, Medicine and Health, University of Manchester, M13 9PT Manchester, UK
| | - Lauren Toms
- Faculty of Biology, Medicine and Health, University of Manchester, M13 9PT Manchester, UK
| | - Graham Coutts
- Faculty of Biology, Medicine and Health, University of Manchester, M13 9PT Manchester, UK
| | - Bernadett Martinecz
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Szigony u. 43, 1083 Budapest, Hungary
| | - Eszter Császár
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Szigony u. 43, 1083 Budapest, Hungary
| | - Gábor Nyiri
- Laboratory of Cerebral Cortex Research, Institute of Experimental Medicine, Szigony u. 43, 1083 Budapest, Hungary
| | - Athina Papaemmanouil
- Faculty of Biology, Medicine and Health, University of Manchester, M13 9PT Manchester, UK
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Werner Müller
- Faculty of Biology, Medicine and Health, University of Manchester, M13 9PT Manchester, UK
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, 23538 Lübeck, Germany
| | - Nancy Rothwell
- Faculty of Biology, Medicine and Health, University of Manchester, M13 9PT Manchester, UK
| | - Sheila Francis
- Department of Infection, Immunity & Cardiovascular Disease, Medical School, University of Sheffield, S10 2RX Sheffield, UK
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, University of Manchester, M13 9PT Manchester, UK
| | - Adam Denés
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Szigony u. 43, 1083 Budapest, Hungary.
| | - Stuart M Allan
- Faculty of Biology, Medicine and Health, University of Manchester, M13 9PT Manchester, UK.
| |
Collapse
|
35
|
Lalu MM, Fergusson DA, Cheng W, Avey MT, Corbett D, Dowlatshahi D, Macleod MR, Sena ES, Moher D, Shorr R, McCann SK, Gray LJ, Hill MD, O'Connor A, Thayer K, Haggar F, Dobriyal A, Chung HS, Welton NJ, Hutton B. Identifying stroke therapeutics from preclinical models: A protocol for a novel application of network meta-analysis. F1000Res 2019; 8:11. [PMID: 30906535 PMCID: PMC6426098 DOI: 10.12688/f1000research.15869.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/08/2018] [Indexed: 12/11/2022] Open
Abstract
Introduction: Globally, stroke is the second leading cause of death. Despite the burden of illness and death, few acute interventions are available to patients with ischemic stroke. Over 1,000 potential neuroprotective therapeutics have been evaluated in preclinical models. It is important to use robust evidence synthesis methods to appropriately assess which therapies should be translated to the clinical setting for evaluation in human studies. This protocol details planned methods to conduct a systematic review to identify and appraise eligible studies and to use a network meta-analysis to synthesize available evidence to answer the following questions: in preclinical in vivo models of focal ischemic stroke, what are the relative benefits of competing therapies tested in combination with the gold standard treatment alteplase in (i) reducing cerebral infarction size, and (ii) improving neurobehavioural outcomes? Methods: We will search Ovid Medline and Embase for articles on the effects of combination therapies with alteplase. Controlled comparison studies of preclinical in vivo models of experimentally induced focal ischemia testing the efficacy of therapies with alteplase versus alteplase alone will be identified. Outcomes to be extracted include infarct size (primary outcome) and neurobehavioural measures. Risk of bias and construct validity will be assessed using tools appropriate for preclinical studies. Here we describe steps undertaken to perform preclinical network meta-analysis to synthesise all evidence for each outcome and obtain a comprehensive ranking of all treatments. This will be a novel use of this evidence synthesis approach in stroke medicine to assess pre-clinical therapeutics. Combining all evidence to simultaneously compare mutliple therapuetics tested preclinically may provide a rationale for the clinical translation of therapeutics for patients with ischemic stroke. Dissemination: Review findings will be submitted to a peer-reviewed journal and presented at relevant scientific meetings to promote knowledge transfer. Registration: PROSPERO number to be submitted following peer review.
Collapse
Affiliation(s)
- Manoj M Lalu
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Ottawa, Canada.,Clinical Epidemiology Program, Blueprint Translational Research Group, The Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada.,Regenerative Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Dean A Fergusson
- Clinical Epidemiology Program, Blueprint Translational Research Group, The Ottawa Hospital Research Institute, Ottawa, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
| | - Wei Cheng
- Knowledge Synthesis Group, Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Marc T Avey
- Clinical Epidemiology Program, Blueprint Translational Research Group, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Dale Corbett
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada.,Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, University of Ottawa, Ottawa, Canada
| | - Dar Dowlatshahi
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada.,Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, University of Ottawa, Ottawa, Canada.,Department of Medicine. Division of Neurology, The Ottawa Hospital, Ottawa, Canada.,Neuroscience Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Malcolm R Macleod
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Emily S Sena
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - David Moher
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada.,Knowledge Synthesis Group, Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Risa Shorr
- Learning Services, The Ottawa Hospital, Ottawa, Canada
| | - Sarah K McCann
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Laura J Gray
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Michael D Hill
- Cumming School of Medicine, Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Annette O'Connor
- College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Kristina Thayer
- National Institutes of Environmental Health Sciences, Durham, North Carolina, USA
| | - Fatima Haggar
- Clinical Epidemiology Program, Blueprint Translational Research Group, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Aditi Dobriyal
- Clinical Epidemiology Program, Blueprint Translational Research Group, The Ottawa Hospital Research Institute, Ottawa, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Hee Sahng Chung
- Clinical Epidemiology Program, Blueprint Translational Research Group, The Ottawa Hospital Research Institute, Ottawa, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Nicky J Welton
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Brian Hutton
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada.,Knowledge Synthesis Group, Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| |
Collapse
|
36
|
Abstract
Any given research claim can be made with a degree of confidence that a phenomenon is present, with an estimate of the precision of the observed effects and a prediction of the extent to which the findings might hold true under different experimental or real-world conditions. In some situations, the certainty and precision obtained from a single study are sufficient reliably to inform future research decisions. However, in other situations greater certainty is required. This might be the case where a substantial research investment is planned, a pivotal claim is to be made or the launch of a clinical trial programme is being considered. Under these circumstances, some form of summary of findings across studies may be helpful.Summary estimates can describe findings from exploratory (observational) or hypothesis testing experiments, but importantly, the creation of such summaries is, in itself, observational rather than experimental research. The process is therefore particularly at risk from selective identification of literature to be included, and this can be addressed using systematic search strategies and pre-specified criteria for inclusion and exclusion against which possible contributing data will be assessed. This characterises a systematic review (in contrast to nonsystematic or narrative reviews). In meta-analysis, there is an attempt to provide a quantitative summary of such research findings.
Collapse
Affiliation(s)
- Malcolm R Macleod
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.
| | | | - Kaitlyn Hair
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Emily Sena
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
37
|
Hadley G, Beard DJ, Couch Y, Neuhaus AA, Adriaanse BA, DeLuca GC, Sutherland BA, Buchan AM. Rapamycin in ischemic stroke: Old drug, new tricks? J Cereb Blood Flow Metab 2019; 39:20-35. [PMID: 30334673 PMCID: PMC6311672 DOI: 10.1177/0271678x18807309] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/16/2018] [Accepted: 09/06/2018] [Indexed: 12/19/2022]
Abstract
The significant morbidity that accompanies stroke makes it one of the world's most devastating neurological disorders. Currently, proven effective therapies have been limited to thrombolysis and thrombectomy. The window for the administration of these therapies is narrow, hampered by the necessity of rapidly imaging patients. A therapy that could extend this window by protecting neurons may improve outcome. Endogenous neuroprotection has been shown to be, in part, due to changes in mTOR signalling pathways and the instigation of productive autophagy. Inducing this effect pharmacologically could improve clinical outcomes. One such therapy already in use in transplant medicine is the mTOR inhibitor rapamycin. Recent evidence suggests that rapamycin is neuroprotective, not only via neuronal autophagy but also through its broader effects on other cells of the neurovascular unit. This review highlights the potential use of rapamycin as a multimodal therapy, acting on the blood-brain barrier, cerebral blood flow and inflammation, as well as directly on neurons. There is significant potential in applying this old drug in new ways to improve functional outcomes for patients after stroke.
Collapse
Affiliation(s)
- Gina Hadley
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Daniel J Beard
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Yvonne Couch
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Ain A Neuhaus
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Bryan A Adriaanse
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Brad A Sutherland
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Alastair M Buchan
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Acute Vascular Imaging Centre, University of Oxford, Oxford University Hospitals, Oxford, UK
| |
Collapse
|
38
|
Xu W, Gao L, Li T, Shao A, Zhang J. Neuroprotective Role of Agmatine in Neurological Diseases. Curr Neuropharmacol 2018; 16:1296-1305. [PMID: 28786346 PMCID: PMC6251039 DOI: 10.2174/1570159x15666170808120633] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 07/17/2017] [Accepted: 07/27/2017] [Indexed: 12/31/2022] Open
Abstract
Background: Neurological diseases have always been one of the leading cause of mobility and mortality world-widely. However, it is still lacking efficient agents. Agmatine, an endogenous polyamine, exerts its diverse biological characteristics and therapeutic potential in varied aspects. Methods: This review would focus on the neuroprotective actions of agmatine and its potential mechanisms in the setting of neurological diseases. Results: Numerous studies had demonstrated the neuroprotective effect of agmatine in varied types of neurological diseases, including acute attack (stroke and trauma brain injury) and chronic neurodegenerative diseases (Parkinson's disease, Alz-heimer’s disease). The potential mechanism of agmatine induced neuroprotection includes anti-oxidation, anti-apoptosis, anti-inflammation, brain blood barrier (BBB) protection and brain edema prevention. Conclusions: The safety and low incidence of adverse effects indicate the vast potential therapeutic value of agmatine in the treatment of neurological diseases. However, most of the available studies relate to the agmatine are conducted in experi-mental models, more clinical trials are needed before the agmatine could be extensively clinically used
Collapse
Affiliation(s)
- Weilin Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Liansheng Gao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tao Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
39
|
Gasdermin Family: a Promising Therapeutic Target for Stroke. Transl Stroke Res 2018; 9:555-563. [DOI: 10.1007/s12975-018-0666-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/17/2018] [Accepted: 09/21/2018] [Indexed: 12/17/2022]
|
40
|
Slevin M, Iemma RS, Zeinolabediny Y, Liu D, Ferris GR, Caprio V, Phillips N, Di Napoli M, Guo B, Zeng X, AlBaradie R, Binsaleh NK, McDowell G, Fang WH. Acetylcholine Inhibits Monomeric C-Reactive Protein Induced Inflammation, Endothelial Cell Adhesion, and Platelet Aggregation; A Potential Therapeutic? Front Immunol 2018; 9:2124. [PMID: 30319609 PMCID: PMC6168760 DOI: 10.3389/fimmu.2018.02124] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 08/28/2018] [Indexed: 11/13/2022] Open
Abstract
Objectives: In this study, we examined the possibility of using targeted antibodies and the potential of small molecular therapeutics (acetylcholine, nicotine and tacrine) to block the pro-inflammatory and adhesion-related properties of monomeric C-reactive protein (mCRP). Methods: We used three established models (platelet aggregation assay, endothelial leucocyte binding assay and monocyte inflammation via ELISA and Western blotting) to assess the potential of these therapeutics. Results: The results of this study showed that monocyte induced inflammation (raised tumor necrosis factor-alpha-TNF-α) induced by mCRP was significantly blocked in the presence of acetylcholine and nicotine, whilst tacrine and targeted antibodies (clones 8C10 and 3H12) had less of or no significant effects. Western blotting confirmed the ability of acetylcholine to inhibit mCRP-induced cell signaling phosphorylation of extracellular signal regulated kinase 1/2 (ERK1/2), p38 and nuclear factor-kappa B (NF-κB). There was no evidence of direct binding between small molecules and mCRP. mCRP also induced endothelial cell-monocyte adhesion in a dose dependent fashion, however, both acetylcholine and nicotine as well as targeting antibodies notably inhibited adhesion. Finally, we investigated their effects on mCRP-induced platelet aggregation. All three small molecules significantly attenuated platelet aggregation as did the antibody 8C10, although 3H12 had a weaker effect. Discussion: Acetylcholine and to a lesser extent nicotine show potential for therapeutic inhibition of mCRP-induced inflammation and cell and platelet adhesion. These results highlight the potential of targeted antibodies and small molecule therapeutics to inhibit the binding of mCRP by prevention of membrane interaction and subsequent activation of cellular cascade systems, which produce the pro-inflammatory effects associated with mCRP.
Collapse
Affiliation(s)
- Mark Slevin
- Faculty of Science and Engineering, School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom.,Institute of Dementia and Neurolgical Aging, Weifang Medical University, Weifang, China.,University of Medicine and Pharmacy, Târgu Mures, Romania
| | - Rocco S Iemma
- Faculty of Science and Engineering, School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
| | - Yasmin Zeinolabediny
- Faculty of Science and Engineering, School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom.,Applied Medical Sciences College, Majmaah University, Al Majma'ah, Saudi Arabia
| | - Donghui Liu
- Faculty of Science and Engineering, School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom.,Applied Medical Sciences College, Majmaah University, Al Majma'ah, Saudi Arabia
| | - Glenn R Ferris
- Faculty of Science and Engineering, School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
| | - Vittorio Caprio
- Faculty of Science and Engineering, School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
| | - Nicola Phillips
- Faculty of Science and Engineering, School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
| | - Mario Di Napoli
- Neurological Service, Ospedale San Camillo de Lellis, Rieti, Italy
| | - Baoqiang Guo
- Faculty of Science and Engineering, School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom.,Institute of Dementia and Neurolgical Aging, Weifang Medical University, Weifang, China
| | - Xianwei Zeng
- Institute of Dementia and Neurolgical Aging, Weifang Medical University, Weifang, China
| | - Raid AlBaradie
- Applied Medical Sciences College, Majmaah University, Al Majma'ah, Saudi Arabia
| | - Naif K Binsaleh
- Faculty of Science and Engineering, School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
| | - Garry McDowell
- Faculty of Science and Engineering, School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
| | - Wen-Hui Fang
- Faculty of Science and Engineering, School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
| |
Collapse
|
41
|
In diabetic ketoacidosis brain injury including cerebral oedema and infarction is caused by interleukin-1. Med Hypotheses 2018; 121:44-46. [PMID: 30396487 DOI: 10.1016/j.mehy.2018.09.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/31/2018] [Accepted: 09/05/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Brain injury in diabetic ketoacidosis (DKA) is common but under recognized and affects up to 54% of patients with this complication. It's manifestations include cerebral oedema (CE) and cerebral infarction (CI). The etiology of CE in DKA has up to the present time been uncertain. Practical management had been guided by the assumption that rapid osmotic shifts due to rapid correction of hypovolemia and reduction of plasma glucose could cause a shift of water into the cells. The osmotic effect of glucose can cause inflammation by activation of inflammasomes. Recently it has been recognized that the body is in a pro-inflammatory state during DKA involving interleukin-1 production by inflammasomes. Interleukin-1 has been involved in the pathogenesis of cerebral oedema and CI. HYPOTHESIS In diabetic ketoacidosis brain injury including cerebral oedema and infarction is caused by interleukin-1. CONFIRMATION OF HYPOTHESIS AND IMPLICATIONS Inflammasome activity could be quantified in peripheral blood mononuclear cells and in patients with and without clinical and/or subclinical CE and/or stroke or features of cerebral ischemia on MRI. Surrogates of brain injury in peripheral blood like neuron specific enolase could be measured and correlated with inflammasome activity. Interleukin-1 receptor antagonists and inflammasome inhibitors including telmisartan could be assessed in their effect on MRI changes consistent with CE or CI in patients with DKA in randomised placebo-controlled trials.
Collapse
|
42
|
Malone K, Amu S, Moore AC, Waeber C. The immune system and stroke: from current targets to future therapy. Immunol Cell Biol 2018; 97:5-16. [DOI: 10.1111/imcb.12191] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/11/2018] [Accepted: 07/16/2018] [Indexed: 01/15/2023]
Affiliation(s)
- Kyle Malone
- Department of Pharmacology and Therapeutics; School of Pharmacy; University College Cork; Cork Ireland
| | - Sylvie Amu
- Department of Pharmacology and Therapeutics; School of Pharmacy; University College Cork; Cork Ireland
| | - Anne C Moore
- Department of Pharmacology and Therapeutics; School of Pharmacy; University College Cork; Cork Ireland
| | - Christian Waeber
- Department of Pharmacology and Therapeutics; School of Pharmacy; University College Cork; Cork Ireland
| |
Collapse
|
43
|
Jiang RH, Wu CJ, Xu XQ, Lu SS, Zu QQ, Zhao LB, Wang J, Liu S, Shi HB. Hypoxic conditioned medium derived from bone marrow mesenchymal stromal cells protects against ischemic stroke in rats. J Cell Physiol 2018; 234:1354-1368. [PMID: 30076722 DOI: 10.1002/jcp.26931] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/12/2018] [Indexed: 12/13/2022]
Abstract
In recent years, studies have shown that the secretome of bone marrow mesenchymal stromal cells (BMSCs) contains many growth factors, cytokines, and antioxidants, which may provide novel approaches to treat ischemic diseases. Furthermore, the secretome may be modulated by hypoxic preconditioning. We hypothesized that conditioned medium (CM) derived from BMSCs plays a crucial role in reducing tissue damage and improving neurological recovery after ischemic stroke and that hypoxic preconditioning of BMSCs robustly improves these activities. Rats were subjected to ischemic stroke by middle cerebral artery occlusion and then intravenously administered hypoxic CM, normoxic CM, or Dulbecco modified Eagle medium (DMEM, control). Cytokine antibody arrays and label-free quantitative proteomics analysis were used to compare the differences between hypoxic CM and normoxic CM. Injection of normoxic CM significantly reduced the infarct area and improved neurological recovery after stroke compared with administering DMEM. These outcomes may be associated with the attenuation of apoptosis and promotion of angiogenesis. Hypoxic preconditioning significantly enhanced these therapeutic effects. Fourteen proteins were significantly increased in hypoxic CM compared with normoxic CM as measured by cytokine arrays. The label-free quantitative proteomics analysis revealed 163 proteins that were differentially expressed between the two groups, including 107 upregulated proteins and 56 downregulated proteins. Collectively, our results demonstrate that hypoxic CM protected brain tissue from ischemic injury and promoted functional recovery after stroke in rats and that hypoxic CM may be the basis of a potential therapy for stroke patients.
Collapse
Affiliation(s)
- Run-Hao Jiang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chen-Jiang Wu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao-Quan Xu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shan-Shan Lu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qing-Quan Zu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lin-Bo Zhao
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Wang
- Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Sheng Liu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hai-Bin Shi
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
44
|
The Pathogenesis of Necroptosis-Dependent Signaling Pathway in Cerebral Ischemic Disease. Behav Neurol 2018; 2018:6814393. [PMID: 30140326 PMCID: PMC6081565 DOI: 10.1155/2018/6814393] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/13/2018] [Indexed: 11/18/2022] Open
Abstract
Necroptosis is the best-described form of regulated necrosis at present, which is widely recognized as a component of caspase-independent cell death mediated by the concerted action of receptor-interacting protein kinase 1 (RIPK1) and receptor-interacting protein kinase 3 (RIPK3). Mixed-lineage kinase domain-like (MLKL) was phosphorylated by RIPK3 at the threonine 357 and serine 358 residues and then formed tetramers and translocated onto the plasma membrane, which destabilizes plasma membrane integrity leading to cell swelling and membrane rupture. Necroptosis is downstream of the tumor necrosis factor (TNF) receptor family, and also interaction with NOD-like receptor pyrin 3 (NLRP3) induced inflammasome activation. Multiple inhibitors of RIPK1 and MLKL have been developed to block the cascade of signal pathways for procedural necrosis and represent potential leads for drug development. In this review, we highlight recent progress in the study of roles for necroptosis in cerebral ischemic disease and discuss how these modifications delicately control necroptosis.
Collapse
|
45
|
Swanton T, Cook J, Beswick JA, Freeman S, Lawrence CB, Brough D. Is Targeting the Inflammasome a Way Forward for Neuroscience Drug Discovery? SLAS DISCOVERY 2018; 23:991-1017. [PMID: 29969573 DOI: 10.1177/2472555218786210] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuroinflammation is becoming increasingly recognized as a critical factor in the pathology of both acute and chronic neurological conditions. Inflammasomes such as the one formed by NACHT, LRR, and PYD domains containing protein 3 (NLRP3) are key regulators of inflammation due to their ability to induce the processing and secretion of interleukin 1β (IL-1β). IL-1β has previously been identified as a potential therapeutic target in a variety of conditions due to its ability to promote neuronal damage under conditions of injury. Thus, inflammasome inhibition has the potential to curtail inflammatory signaling, which could prove beneficial in certain diseases. In this review, we discuss the evidence for inflammasome contributions to the pathology of neurodegenerative conditions such as Alzheimer's disease and Parkinson's disease, epilepsy, and acute degeneration following brain trauma or stroke. In addition, we review the current landscape of drug development targeting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Tessa Swanton
- 1 Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - James Cook
- 1 Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - James A Beswick
- 2 Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Sally Freeman
- 2 Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Catherine B Lawrence
- 1 Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - David Brough
- 1 Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| |
Collapse
|
46
|
Smith CJ, Hulme S, Vail A, Heal C, Parry-Jones AR, Scarth S, Hopkins K, Hoadley M, Allan SM, Rothwell NJ, Hopkins SJ, Tyrrell PJ. SCIL-STROKE (Subcutaneous Interleukin-1 Receptor Antagonist in Ischemic Stroke): A Randomized Controlled Phase 2 Trial. Stroke 2018; 49:1210-1216. [PMID: 29567761 DOI: 10.1161/strokeaha.118.020750] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND PURPOSE The proinflammatory cytokine IL-1 (interleukin-1) has a deleterious role in cerebral ischemia, which is attenuated by IL-1 receptor antagonist (IL-1Ra). IL-1 induces peripheral inflammatory mediators, such as interleukin-6, which are associated with worse prognosis after ischemic stroke. We investigated whether subcutaneous IL-1Ra reduces the peripheral inflammatory response in acute ischemic stroke. METHODS SCIL-STROKE (Subcutaneous Interleukin-1 Receptor Antagonist in Ischemic Stroke) was a single-center, double-blind, randomized, placebo-controlled phase 2 trial of subcutaneous IL-1Ra (100 mg administered twice daily for 3 days) in patients presenting within 5 hours of ischemic stroke onset. Randomization was stratified for baseline National Institutes of Health Stroke Scale score and thrombolysis. Measurement of plasma interleukin-6 and other peripheral inflammatory markers was undertaken at 5 time points. The primary outcome was difference in concentration of log(interleukin-6) as area under the curve to day 3. Secondary outcomes included exploratory effect of IL-1Ra on 3-month outcome with the modified Rankin Scale. RESULTS We recruited 80 patients (mean age, 72 years; median National Institutes of Health Stroke Scale, 12) of whom 73% received intravenous thrombolysis with alteplase. IL-1Ra significantly reduced plasma interleukin-6 (P<0.001) and plasma C-reactive protein (P<0.001). IL-1Ra was well tolerated with no safety concerns. Allocation to IL-1Ra was not associated with a favorable outcome on modified Rankin Scale: odds ratio (95% confidence interval)=0.67 (0.29-1.52), P=0.34. Exploratory mediation analysis suggested that IL-1Ra improved clinical outcome by reducing inflammation, but there was a statistically significant, alternative mechanism countering this benefit. CONCLUSIONS IL-1Ra reduced plasma inflammatory markers which are known to be associated with worse clinical outcome in ischemic stroke. Subcutaneous IL-1Ra is safe and well tolerated. Further experimental studies are required to investigate efficacy and possible interactions of IL-1Ra with thrombolysis. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: ISRCTN74236229.
Collapse
Affiliation(s)
- Craig J Smith
- From the Greater Manchester Comprehensive Stroke Centre, Manchester Academic Health Science Centre, Salford Royal NHS Foundation Trust, United Kingdom (C.J.S., A.R.P.-J., P.J.T.)
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
| | - Sharon Hulme
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
| | - Andy Vail
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
- Centre for Biostatistics, University of Manchester, Manchester Academic Health Science Centre, United Kingdom (A.V., C.H.)
| | - Calvin Heal
- Centre for Biostatistics, University of Manchester, Manchester Academic Health Science Centre, United Kingdom (A.V., C.H.)
| | - Adrian R Parry-Jones
- From the Greater Manchester Comprehensive Stroke Centre, Manchester Academic Health Science Centre, Salford Royal NHS Foundation Trust, United Kingdom (C.J.S., A.R.P.-J., P.J.T.)
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
| | - Sylvia Scarth
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
| | - Karen Hopkins
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
| | - Margaret Hoadley
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
| | - Stuart M Allan
- Division of Neuroscience and Experimental Psychology, University of Manchester, United Kingdom (S.M.A., N.J.R.)
| | - Nancy J Rothwell
- Division of Neuroscience and Experimental Psychology, University of Manchester, United Kingdom (S.M.A., N.J.R.)
| | - Stephen J Hopkins
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
| | - Pippa J Tyrrell
- From the Greater Manchester Comprehensive Stroke Centre, Manchester Academic Health Science Centre, Salford Royal NHS Foundation Trust, United Kingdom (C.J.S., A.R.P.-J., P.J.T.)
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (C.J.S., S.H., A.R.P.-J., S.S., K.H., M.H., S.J.H., P.J.T.)
| |
Collapse
|
47
|
Liu X, Quan N. Microglia and CNS Interleukin-1: Beyond Immunological Concepts. Front Neurol 2018; 9:8. [PMID: 29410649 PMCID: PMC5787061 DOI: 10.3389/fneur.2018.00008] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/05/2018] [Indexed: 12/12/2022] Open
Abstract
Activation of microglia and expression of the inflammatory cytokine interleukin-1 (IL-1) in the CNS have become almost synonymous with neuroinflammation. In numerous studies, increased CNS IL-1 expression and altered microglial morphology have been used as hallmarks of CNS inflammation. A central concept of how CNS IL-1 and microglia influence functions of the nervous system was derived from the notion initially generated in the peripheral immune system: IL-1 stimulates monocyte/macrophage (the peripheral counterparts of microglia) to amplify inflammation. It is increasingly clear, however, CNS IL-1 acts on other targets in the CNS and microglia participates in many neural functions that are not related to immunological activities. Further, CNS exhibits immunological privilege (although not as absolute as previously thought), rendering amplification of inflammation within CNS under stringent control. This review will analyze current literature to evaluate the contribution of immunological and non-immunological aspects of microglia/IL-1 interaction in the CNS to gain insights for how these aspects might affect health and disease in the nervous tissue.
Collapse
Affiliation(s)
- Xiaoyu Liu
- College of Medicine, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| | - Ning Quan
- College of Medicine, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States.,Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
48
|
Ma J, Sun X, Guo T, Su H, Chen Q, Gong Z, Qi J, Zhao X. Interleukin-1 receptor antagonist inhibits angiogenesis via blockage IL-1α/PI3K/NF-κβ pathway in human colon cancer cell. Cancer Manag Res 2017; 9:481-493. [PMID: 29062241 PMCID: PMC5640395 DOI: 10.2147/cmar.s147699] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE This article investigates the relationship between cancer cells and stromal cells in carcinoma cell living microenvironment and elucidates the mechanism that interleukin-1 receptor antagonist (IL-1RA) blocks metastatic potential in colon cancer. METHODS Western blot and RT-PCR assay were used to determine the expression of hepatocyte growth factor (HGF) and IL-1α in colon carcinoma cells and stromal cells. Effect of IL-1RA and HGF on metastatic potential of colon cancer cells were examined by proliferation, invasion, and angiogenesis assays. The interactional role of IL-1RA and HGF were detected by ELISA assay, invasion, and angiogenesis assay making up a co-culture system consisting of stromal and colon cancer cells in cells living microenvironment. RESULTS IL-1α was expressed in human umbilical vein endothelial cells (HUVECs) and HT-29 and WiDr (colon cancer cell lines with higher liver metastatic potential). HGF was expressed only in fibroblast. HGF secretion from fibroblasts was significantly inhibited by IL-1RA (P<0.01). Furthermore, IL-1RA could significantly inhibit migration, proliferation, and angiogenesis of HUVECs (P<0.01). In the double co-culture system, there is a high liver metastatic potential of colon cancer cell line (HT-29) because it can secrete autocrine IL-1α, significantly enhanced angiogenesis compared with low liver metastatic cell line (CaCo-2) (P<0.01), which does not secrete IL-1α. On the contrary, blockage of autocrine IL-1α by IL-1RA might significantly decrease metastatic potential of colon carcinoma cells through downregulation of IL-1α/PI3K/NF-κB pathway. CONCLUSION IL-1 receptor antagonist (IL-1RA) is an important inhibitor in metastatic process of colon carcinoma cell. Based on the above results, we suggest that IL-1RA may be a promising new therapeutic approach in inhibiting colon cancer with IL-1-producing patients.
Collapse
Affiliation(s)
- Jiachi Ma
- Department of General Surgery, Gansu Provincial People’s Hospital, LanZhou
| | - Xiaowen Sun
- Department of Dermatology, The First Hospital of Tianshui, Tianshui
| | - Tiankang Guo
- Department of General Surgery, Gansu Provincial People’s Hospital, LanZhou
| | - He Su
- Department of General Surgery, Gansu Provincial People’s Hospital, LanZhou
| | - Quan Chen
- Department of General Surgery, Gansu Provincial People’s Hospital, LanZhou
| | - Zhenqiang Gong
- Ningxia Medical University Graduate School of Medical Sciences, Surgical Oncology, Yinchuan, People’s Republic of China
| | - Jianbo Qi
- Ningxia Medical University Graduate School of Medical Sciences, Surgical Oncology, Yinchuan, People’s Republic of China
| | - Xiaodan Zhao
- Ningxia Medical University Graduate School of Medical Sciences, Surgical Oncology, Yinchuan, People’s Republic of China
| |
Collapse
|
49
|
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: 93] [Impact Index Per Article: 11.6] [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
|
50
|
Gandolfi M, Smania N, Vella A, Picelli A, Chirumbolo S. Assessed and Emerging Biomarkers in Stroke and Training-Mediated Stroke Recovery: State of the Art. Neural Plast 2017; 2017:1389475. [PMID: 28373915 PMCID: PMC5360976 DOI: 10.1155/2017/1389475] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/11/2017] [Indexed: 12/13/2022] Open
Abstract
Since the increasing update of the biomolecular scientific literature, biomarkers in stroke have reached an outstanding and remarkable revision in the very recent years. Besides the diagnostic and prognostic role of some inflammatory markers, many further molecules and biological factors have been added to the list, including tissue derived cytokines, growth factor-like molecules, hormones, and microRNAs. The literatures on brain derived growth factor and other neuroimmune mediators, bone-skeletal muscle biomarkers, cellular and immunity biomarkers, and the role of microRNAs in stroke recovery were reviewed. To date, biomarkers represent a possible challenge in the diagnostic and prognostic evaluation of stroke onset, pathogenesis, and recovery. Many molecules are still under investigation and may become promising and encouraging biomarkers. Experimental and clinical research should increase this list and promote new discoveries in this field, to improve stroke diagnosis and treatment.
Collapse
Affiliation(s)
- Marialuisa Gandolfi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- UOC Neurorehabilitation, AOUI Verona, Verona, Italy
| | - Nicola Smania
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- UOC Neurorehabilitation, AOUI Verona, Verona, Italy
| | - Antonio Vella
- Immunology Unit, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Alessandro Picelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- UOC Neurorehabilitation, AOUI Verona, Verona, Italy
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|