1
|
Hanin A, Muscal E, Hirsch LJ. Second-line immunotherapy in new onset refractory status epilepticus. Epilepsia 2024; 65:1203-1223. [PMID: 38430119 DOI: 10.1111/epi.17933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/06/2024] [Accepted: 02/12/2024] [Indexed: 03/03/2024]
Abstract
Several pieces of evidence suggest immune dysregulation could trigger the onset and modulate sequelae of new onset refractory status epilepticus (NORSE), including its subtype with prior fever known as febrile infection-related epilepsy syndrome (FIRES). Consensus-driven recommendations have been established to guide the initiation of first- and second-line immunotherapies in these patients. Here, we review the literature to date on second-line immunotherapy for NORSE/FIRES, presenting results from 28 case reports and series describing the use of anakinra, tocilizumab, or intrathecal dexamethasone in 75 patients with NORSE. Among them, 52 patients were managed with anakinra, 21 with tocilizumab, and eight with intrathecal dexamethasone. Most had elevated serum or cerebrospinal fluid cytokine levels at treatment initiation. Treatments were predominantly initiated during the acute phase of the disease (92%) and resulted, within the first 2 weeks, in seizure control for up to 73% of patients with anakinra, 70% with tocilizumab, and 50% with intrathecal dexamethasone. Cytokine levels decreased after treatment for most patients. Anakinra and intrathecal dexamethasone were mainly initiated in children with FIRES, whereas tocilizumab was more frequently prescribed for adults, with or without a prior febrile infection. There was no clear correlation between the response to treatment and the time to initiate the treatment. Most patients experienced long-term disability and drug-resistant post-NORSE epilepsy. Initiation of second-line immunotherapies during status epilepticus (SE) had no clear effect on the emergence of post-NORSE epilepsy or long-term functional outcomes. In a small number of cases, the initiation of anakinra or tocilizumab several years after SE onset resulted in a reduction of seizure frequency for 67% of patients. These data highlight the potential utility of anakinra, tocilizumab, and intrathecal dexamethasone in patients with NORSE. There continues to be interest in the utilization of early cytokine measurements to guide treatment selection and response. Prospective studies are necessary to understand the role of early immunomodulation and its associations with epilepsy and functional outcomes.
Collapse
Affiliation(s)
- Aurélie Hanin
- Comprehensive Epilepsy Center, Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, Inserm, CNRS, Assistance Publique - Hôpitaux de Paris, Hôpital de la Pitié-Salpêtrière, Paris, France
- Epilepsy Unit and Clinical Neurophysiology Department, DMU Neurosciences 6, Assistance Publique - Hôpitaux de Paris, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Eyal Muscal
- Department of Pediatrics, Section of Rheumatology, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas, USA
| | - Lawrence J Hirsch
- Comprehensive Epilepsy Center, Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
2
|
Noll JM, Sherafat AA, Ford GD, Ford BD. The case for neuregulin-1 as a clinical treatment for stroke. Front Cell Neurosci 2024; 18:1325630. [PMID: 38638304 PMCID: PMC11024452 DOI: 10.3389/fncel.2024.1325630] [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: 10/22/2023] [Accepted: 03/01/2024] [Indexed: 04/20/2024] Open
Abstract
Ischemic stroke is the leading cause of serious long-term disability and the 5th leading cause of death in the United States. Revascularization of the occluded cerebral artery, either by thrombolysis or endovascular thrombectomy, is the only effective, clinically-approved stroke therapy. Several potentially neuroprotective agents, including glutamate antagonists, anti-inflammatory compounds and free radical scavenging agents were shown to be effective neuroprotectants in preclinical animal models of brain ischemia. However, these compounds did not demonstrate efficacy in clinical trials with human patients following stroke. Proposed reasons for the translational failure include an insufficient understanding on the cellular and molecular pathophysiology of ischemic stroke, lack of alignment between preclinical and clinical studies and inappropriate design of clinical trials based on the preclinical findings. Therefore, novel neuroprotective treatments must be developed based on a clearer understanding of the complex spatiotemporal mechanisms of ischemic stroke and with proper clinical trial design based on the preclinical findings from specific animal models of stroke. We and others have demonstrated the clinical potential for neuregulin-1 (NRG-1) in preclinical stroke studies. NRG-1 significantly reduced ischemia-induced neuronal death, neuroinflammation and oxidative stress in rodent stroke models with a therapeutic window of >13 h. Clinically, NRG-1 was shown to be safe in human patients and improved cardiac function in multisite phase II studies for heart failure. This review summarizes previous stroke clinical candidates and provides evidence that NRG-1 represents a novel, safe, neuroprotective strategy that has potential therapeutic value in treating individuals after acute ischemic stroke.
Collapse
Affiliation(s)
- Jessica M. Noll
- Division of Biomedical Sciences, University of California-Riverside School of Medicine, Riverside, CA, United States
- Nanostring Technologies, Seattle, WA, United States
| | - Arya A. Sherafat
- Division of Biomedical Sciences, University of California-Riverside School of Medicine, Riverside, CA, United States
| | - Gregory D. Ford
- Southern University-New Orleans, New Orleans, LA, United States
| | - Byron D. Ford
- Department of Anatomy, Howard University College of Medicine, Washington, DC, United States
| |
Collapse
|
3
|
Liu Q, Shi K, Wang Y, Shi FD. Neurovascular Inflammation and Complications of Thrombolysis Therapy in Stroke. Stroke 2023; 54:2688-2697. [PMID: 37675612 DOI: 10.1161/strokeaha.123.044123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Intravenous thrombolysis via tPA (tissue-type plasminogen activator) is the only approved pharmacological treatment for acute ischemic stroke, but its benefits are limited by hemorrhagic transformation. Emerging evidence reveals that tPA swiftly mobilizes immune cells which extravasate into the brain parenchyma via the cerebral vasculature, augmenting neurovascular inflammation, and tissue injury. In this review, we summarize the pronounced alterations of immune cells induced by tPA in patients with stroke and experimental stroke models. We argue that neuroinflammation, triggered by ischemia-induced cell death and exacerbated by tPA, compromises neurovascular integrity and the microcirculation, leading to hemorrhagic transformation. Finally, we discuss current and future approaches to attenuate thrombolysis-associated hemorrhagic transformation via uncoupling immune cells from the neurovascular unit.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Neurology, Tianjin Medical University General Hospital, China (Q.L., F.-D.S.)
| | - Kaibin Shi
- Department of Neurology, National Clinical Research Center for Neurological Diseases of China, Beijing Tiantan Hospital, Capital Medical University (K.S., Y.W., F.-D.S.)
| | - Yongjun Wang
- Department of Neurology, National Clinical Research Center for Neurological Diseases of China, Beijing Tiantan Hospital, Capital Medical University (K.S., Y.W., F.-D.S.)
| | - Fu-Dong Shi
- Department of Neurology, Tianjin Medical University General Hospital, China (Q.L., F.-D.S.)
- Department of Neurology, National Clinical Research Center for Neurological Diseases of China, Beijing Tiantan Hospital, Capital Medical University (K.S., Y.W., F.-D.S.)
| |
Collapse
|
4
|
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]
|
5
|
Inhibiting peripheral and central MAO-B ameliorates joint inflammation and cognitive impairment in rheumatoid arthritis. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1188-1200. [PMID: 35982301 PMCID: PMC9440195 DOI: 10.1038/s12276-022-00830-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 05/07/2022] [Accepted: 05/31/2022] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder characterized by chronic inflammation and the destruction of joints and systemic organs. RA is commonly accompanied by neuropsychiatric complications, such as cognitive impairment and depression. However, the role of monoamine oxidase (MAO) and its inhibitors in controlling neurotransmitters associated with these complications in RA have not been clearly identified. Here, we report that peripheral and central MAO-B are highly associated with joint inflammation and cognitive impairment in RA, respectively. Ribonucleic acid (RNA) sequencing and protein expression quantification were used to show that MAO-B and related molecules, such as gamma aminobutyric acid (GABA), were elevated in the inflamed synovium of RA patients. In primary cultured fibroblast-like synoviocytes in the RA synovium, MAO-B expression was significantly increased by tumor necrosis factor (TNF)-α-induced autophagy, which produces putrescine, the polyamine substrate for GABA synthesis. We also observed that MAO-B-mediated aberrant astrocytic production of GABA was augmented by interleukin (IL)-1β and inhibited CA1-hippocampal pyramidal neurons, which are responsible for memory storage, in an animal model of RA. Moreover, a newly developed reversible inhibitor of MAO-B ameliorated joint inflammation by inhibiting cyclooxygenase (Cox)-2. Therefore, MAO-B can be an effective therapeutic target for joint inflammation and cognitive impairment in patients with RA. Inhibiting an enzyme that is upregulated during joint inflammation may prove a valuable therapy for rheumatoid arthritis (RA). As well as causing considerable pain and discomfort in the joints, RA can also trigger neuropsychiatric problems including depression and memory impairment. The monoamine oxidase (MAO) enzyme family is involved in the control of neurotransmitters, and there is evidence that links MAO-B levels with systemic inflammation. C. Justin Lee at Center for Cognition and Sociality, Institute for Basic Science,, Daejeon, South Korea, and co-workers examined the role of MAO-B in RA using patient tissue samples and mouse models. MAO-B and related molecules were upregulated in patients’ inflamed joint tissues. In mice, elevated MAO-B triggered the inhibition of nerve cell activity related to memory storage. A novel drug that inhibits MAO-B reduced RA-related inflammation and cognitive impairment in mice, suggesting a promising approach to treatment.
Collapse
|
6
|
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
|
7
|
Pradillo JM, Hernández-Jiménez M, Fernández-Valle ME, Medina V, Ortuño JE, Allan SM, Proctor SD, Garcia-Segura JM, Ledesma-Carbayo MJ, Santos A, Moro MA, Lizasoain I. Influence of metabolic syndrome on post-stroke outcome, angiogenesis and vascular function in old rats determined by dynamic contrast enhanced MRI. J Cereb Blood Flow Metab 2021; 41:1692-1706. [PMID: 34152893 PMCID: PMC8221771 DOI: 10.1177/0271678x20976412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Stroke affects primarily aged and co-morbid people, aspects not properly considered to date. Since angiogenesis/vasculogenesis are key processes for stroke recovery, we purposed to determine how different co-morbidities affect the outcome and angiogenesis/vasculogenesis, using a rodent model of metabolic syndrome, and by dynamic enhanced-contrast imaging (DCE-MRI) to assess its non-invasive potential to determine these processes. Twenty/twenty-two month-old corpulent (JCR:LA-Cp/Cp), a model of metabolic syndrome and lean rats were used. After inducing the experimental ischemia by transient MCAO, angiogenesis was analyzed by histology, vasculogenesis by determination of endothelial progenitor cells in peripheral blood by flow cytometry and evaluating their pro-angiogenic properties in culture and the vascular function by DCE-MRI at 3, 7 and 28 days after tMCAO. Our results show an increased infarct volume, BBB damage and an impaired outcome in corpulent rats compared with their lean counterparts. Corpulent rats also displayed worse post-stroke angiogenesis/vasculogenesis, outcome that translated in an impaired vascular function determined by DCE-MRI. These data confirm that outcome and angiogenesis/vasculogenesis induced by stroke in old rats are negatively affected by the co-morbidities present in the corpulent genotype and also that DCE-MRI might be a technique useful for the non-invasive evaluation of vascular function and angiogenesis processes.
Collapse
Affiliation(s)
- Jesús M Pradillo
- Neurovascular Research Unit, Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid and Instituto de Investigación Hospital 12 de Octubre i+12, Madrid, Spain
| | - Macarena Hernández-Jiménez
- Neurovascular Research Unit, Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid and Instituto de Investigación Hospital 12 de Octubre i+12, Madrid, Spain
| | - María E Fernández-Valle
- Neurovascular Research Unit, Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid and Instituto de Investigación Hospital 12 de Octubre i+12, Madrid, Spain
| | - Violeta Medina
- Neurovascular Research Unit, Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid and Instituto de Investigación Hospital 12 de Octubre i+12, Madrid, Spain
| | - Juan E Ortuño
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.,Biomedical Image Technologies (BIT), ETSI Telecomunicación, Universidad Politécnica de Madrid, Spain
| | - Stuart M Allan
- 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
| | - Spencer D Proctor
- Division of Human Nutrition, Metabolic and Cardiovascular Diseases Laboratory, Agricultural, Food and Nutritional Science Li Ka Shing (LKS) Centre for Health Research Innovation, University of Alberta, Edmonton, Canada
| | - Juan M Garcia-Segura
- Neurovascular Research Unit, Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid and Instituto de Investigación Hospital 12 de Octubre i+12, Madrid, Spain
| | - María J Ledesma-Carbayo
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.,Biomedical Image Technologies (BIT), ETSI Telecomunicación, Universidad Politécnica de Madrid, Spain
| | - Andrés Santos
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.,Biomedical Image Technologies (BIT), ETSI Telecomunicación, Universidad Politécnica de Madrid, Spain
| | - María A Moro
- Neurovascular Research Unit, Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid and Instituto de Investigación Hospital 12 de Octubre i+12, Madrid, Spain
| | - Ignacio Lizasoain
- Neurovascular Research Unit, Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid and Instituto de Investigación Hospital 12 de Octubre i+12, Madrid, Spain
| |
Collapse
|
8
|
Badr S, Rezq S, Abdelghany RH. Endogenous β-endorphin plays a pivotal role in angiotensin II-mediated central neurochemical changes and pressor response. Chem Biol Interact 2021; 342:109475. [PMID: 33872574 DOI: 10.1016/j.cbi.2021.109475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/08/2021] [Accepted: 04/12/2021] [Indexed: 10/21/2022]
Abstract
Endorphins are endogenous opioid neuropeptides that are mainly produced from pituitary gland in response to pain and different triggers including interleukin 1 beta (IL-1β) and corticotropin-releasing factor (CRF). Angiotensin II (Ang II) can stimulate β-endorphin production, but the exact molecular mechanisms involved in this effect, and the role of the released β-endorphin in Ang II-mediated pressor response remain elusive. Male rats were injected with IL-1β receptor antagonist (IL-1Ra, 100 μg/kg), the CRF receptor blocker, astressin (20 μg/rat) or a combination of both, prior to Ang II injection (200 μg/kg). Another group of rats was given naloxone (1.6 mg/kg) or telmisartan (5 mg/kg) before Ang II injection. Blood pressure and serum and Paraventricular nucleus (PVN) β-endorphin were detected. Moreover, IL-1β and CRF as well as markers of oxidative stress [malondialdehyde (MDA) and superoxide dismutase (SOD)], inflammation [C-reactive protein (CRP)] and neuronal activation (c-Fos, l-glutamate, and phosphorylated ERK) were measured in the PVN of different groups. Ang II induced a pressor response and increased serum and PVN β-endorphin levels that were attenuated in rats pre-treated with astressin or/and IL-1Ra. Moreover, Ang II increased PVN oxidative stress, inflammation and neuronal activation. Telmisartan abolished the previous effects, while naloxone, astressin and IL-1Ra aggravated Ang II-mediated pressor response and most of the biochemical changes. These findings suggest that, Ang II can induce β-endorphin release via increasing both IL-1β and CRF levels which in result mitigates Ang II-mediated central responses. This study highlights β-endorphin as a possible target for treating hypertension.
Collapse
Affiliation(s)
- Safy Badr
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Samar Rezq
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt.
| | - Rasha H Abdelghany
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
9
|
Yates AG, Jogia T, Gillespie ER, Couch Y, Ruitenberg MJ, Anthony DC. Acute IL-1RA treatment suppresses the peripheral and central inflammatory response to spinal cord injury. J Neuroinflammation 2021; 18:15. [PMID: 33407641 PMCID: PMC7788822 DOI: 10.1186/s12974-020-02050-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/09/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The acute phase response (APR) to CNS insults contributes to the overall magnitude and nature of the systemic inflammatory response. Aspects of this response are thought to drive secondary inflammatory pathology at the lesion site, and suppression of the APR can therefore afford some neuroprotection. In this study, we examined the APR in a mouse model of traumatic spinal cord injury (SCI), along with its relationship to neutrophil recruitment during the immediate aftermath of the insult. We specifically investigated the effect of IL-1 receptor antagonist (IL-1RA) administration on the APR and leukocyte recruitment to the injured spinal cord. METHODS Adult female C57BL/6 mice underwent either a 70kD contusive SCI, or sham surgery, and tissue was collected at 2, 6, 12, and 24 hours post-operation. For IL-1RA experiments, SCI mice received two intraperitoneal injections of human IL-1RA (100mg/kg), or saline as control, immediately following, and 5 hours after impact, and animals were sacrificed 6 hours later. Blood, spleen, liver and spinal cord were collected to study markers of central and peripheral inflammation by flow cytometry, immunohistochemistry and qPCR. Results were analysed by two-way ANOVA or student's t-test, as appropriate. RESULTS SCI induced a robust APR, hallmarked by elevated hepatic expression of pro-inflammatory marker genes and a significantly increased neutrophil presence in the blood, liver and spleen of these animals, as early as 2 hours after injury. This peripheral response preceded significant neutrophil infiltration of the spinal cord, which peaked 24 hours post-SCI. Although expression of IL-1RA was also induced in the liver following SCI, its response was delayed compared to IL-1β. Exogenous administration of IL-1RA during this putative therapeutic window was able to suppress the hepatic APR, as evidenced by a reduction in CXCL1 and SAA-2 expression as well as a significant decrease in neutrophil infiltration in both the liver and the injured spinal cord itself. CONCLUSIONS Our data indicate that peripheral administration of IL-1RA can attenuate the APR which in turn reduces immune cell infiltration at the spinal cord lesion site. We propose IL-1RA treatment as a viable therapeutic strategy to minimise the harmful effects of SCI-induced inflammation.
Collapse
Affiliation(s)
- Abi G Yates
- Department of Pharmacology, The University of Oxford, Mansfield Road, Oxford, UK
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Queensland, Australia
| | - Trisha Jogia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Queensland, Australia
| | - Ellen R Gillespie
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Queensland, Australia
| | - Yvonne Couch
- Acute Stroke Programme, RDM-Investigative Medicine, The University of Oxford, Oxford, UK
| | - Marc J Ruitenberg
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Queensland, Australia
| | - Daniel C Anthony
- Department of Pharmacology, The University of Oxford, Mansfield Road, Oxford, UK.
- Sechenov First Moscow State Medical University, Moscow, Russia.
| |
Collapse
|
10
|
Dyomina AV, Zubareva OE, Smolensky IV, Vasilev DS, Zakharova MV, Kovalenko AA, Schwarz AP, Ischenko AM, Zaitsev AV. Anakinra Reduces Epileptogenesis, Provides Neuroprotection, and Attenuates Behavioral Impairments in Rats in the Lithium-Pilocarpine Model of Epilepsy. Pharmaceuticals (Basel) 2020; 13:ph13110340. [PMID: 33113868 PMCID: PMC7692198 DOI: 10.3390/ph13110340] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/18/2020] [Accepted: 10/22/2020] [Indexed: 12/17/2022] Open
Abstract
Temporal lobe epilepsy is a widespread chronic disorder that manifests as spontaneous seizures and is often characterized by refractoriness to drug treatment. Temporal lobe epilepsy can be caused by a primary brain injury; therefore, the prevention of epileptogenesis after a primary event is considered one of the best treatment options. However, a preventive treatment for epilepsy still does not exist. Neuroinflammation is directly involved in epileptogenesis and neurodegeneration, leading to the epileptic condition and cognitive decline. In the present study, we aimed to clarify the effect of treatment with a recombinant form of the Interleukin-1 receptor antagonist (anakinra) on epileptogenesis and behavioral impairments in rats using the lithium–pilocarpine model. We found that anakinra administration during the latent phase of the model significantly suppressed the duration and frequency of spontaneous recurrent seizures in the chronic phase. Moreover, anakinra administration prevented some behavioral impairments, including motor hyperactivity and disturbances in social interactions, during both the latent and chronic periods. Histological analysis revealed that anakinra administration decreased neuronal loss in the CA1 and CA3 areas of the hippocampus but did not prevent astro- and microgliosis. The treatment increased the expression level of the solute carrier family 1 member 2 gene (Slc1a2, encoding excitatory amino acid transporter 2 (EAAT2)) in the hippocampus, potentially leading to a neuroprotective effect. However, the increased gene expression of proinflammatory cytokine genes (Interleukin-1β (Il1b) and tumor necrosis factor α (Tnfa)) and astroglial marker genes (glial fibrillary acidic protein (Gfap) and inositol 1,4,5-trisphosphate receptor type 2 (Itpr2)) in experimental rats was not affected by anakinra treatment. Thus, our data demonstrate that the administration of anakinra during epileptogenesis has some beneficial disease-modifying effects.
Collapse
Affiliation(s)
- Alexandra V. Dyomina
- Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, 44, Toreza Prospekt, 194223 Saint Petersburg, Russia; (A.V.D.); (O.E.Z.); (I.V.S.); (D.S.V.) (M.V.Z.); (A.A.K.); (A.P.S.)
| | - Olga E. Zubareva
- Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, 44, Toreza Prospekt, 194223 Saint Petersburg, Russia; (A.V.D.); (O.E.Z.); (I.V.S.); (D.S.V.) (M.V.Z.); (A.A.K.); (A.P.S.)
| | - Ilya V. Smolensky
- Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, 44, Toreza Prospekt, 194223 Saint Petersburg, Russia; (A.V.D.); (O.E.Z.); (I.V.S.); (D.S.V.) (M.V.Z.); (A.A.K.); (A.P.S.)
| | - Dmitry S. Vasilev
- Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, 44, Toreza Prospekt, 194223 Saint Petersburg, Russia; (A.V.D.); (O.E.Z.); (I.V.S.); (D.S.V.) (M.V.Z.); (A.A.K.); (A.P.S.)
| | - Maria V. Zakharova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, 44, Toreza Prospekt, 194223 Saint Petersburg, Russia; (A.V.D.); (O.E.Z.); (I.V.S.); (D.S.V.) (M.V.Z.); (A.A.K.); (A.P.S.)
| | - Anna A. Kovalenko
- Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, 44, Toreza Prospekt, 194223 Saint Petersburg, Russia; (A.V.D.); (O.E.Z.); (I.V.S.); (D.S.V.) (M.V.Z.); (A.A.K.); (A.P.S.)
| | - Alexander P. Schwarz
- Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, 44, Toreza Prospekt, 194223 Saint Petersburg, Russia; (A.V.D.); (O.E.Z.); (I.V.S.); (D.S.V.) (M.V.Z.); (A.A.K.); (A.P.S.)
| | - Alexander M. Ischenko
- Research Institute of Highly Pure Biopreparations, Federal Medical-Biological Agency, 7, Pudozhskaya Street, 197110 Saint Petersburg, Russia;
| | - Aleksey V. Zaitsev
- Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, 44, Toreza Prospekt, 194223 Saint Petersburg, Russia; (A.V.D.); (O.E.Z.); (I.V.S.); (D.S.V.) (M.V.Z.); (A.A.K.); (A.P.S.)
- Correspondence: ; Tel.: +7-812-552-3058
| |
Collapse
|
11
|
Zhang H, Wei M, Lu X, Sun Q, Wang C, Zhang J, Fan H. Aluminum trichloride caused hippocampal neural cells death and subsequent depression-like behavior in rats via the activation of IL-1β/JNK signaling pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 715:136942. [PMID: 32007895 DOI: 10.1016/j.scitotenv.2020.136942] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/20/2020] [Accepted: 01/24/2020] [Indexed: 06/10/2023]
Abstract
Aluminum (Al) is an inorganic pollutant that induces nerve cells apoptosis and necroptosis, thereby causing depression and neurodegenerative diseases. IL-1β/JNK signaling pathway can regulate apoptosis and necroptosis. However, it remains unclear whether IL-1β/JNK signaling pathway is involving in the regulation of Al-induced hippocampal neural cells apoptosis and necroptosis. To investigate the mechanism of Al on neural cells apoptosis and necroptosis, rats were orally exposed to different doses of AlCl3 for 90 days. The open-field test results showed that AlCl3 caused depressive behavior in rats. Histopathological evidence showed that AlCl3 induced hippocampal neural cells apoptosis and necrosis. Moreover, Bax/Bcl-2 mRNA expression ratio, caspase-3 activity and mRNA expression and TUNEL positive rates were upregulated, meanwhile, TNF-α mRNA and protein expression levels, TNFR1, RIP1, RIP3 and MLKL proteins levels were increased, while caspase-8 protein level was decreased in the hippocampus of Al-exposed groups. These results proved that AlCl3 induced hippocampal neural cells apoptosis and necroptosis. Combined with histopathology and correlation analysis, we deduced that hippocampal neural cells were more likely to undergo necroptosis at high doses (450 mg/kg) of AlCl3, while <150 mg/kg AlCl3 tended to induce apoptosis. Finally, AlCl3 increased the proteins level of IL-1β, IL-1RI, IL-1RAcP, JNK and p-JNK, indicating that AlCl3 activated IL-1β/JNK signaling pathway. However, the application of IL-1 receptor antagonist (IL-1Ra) inhibited the phosphorylation of JNK and the related genes expression of apoptosis and necroptosis caused by AlCl3. Thus, we concluded that AlCl3 induced hippocampal neural cells death and depression-like behavior in rats by activating IL-1β/JNK signaling pathway.
Collapse
Affiliation(s)
- Haiyang Zhang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Mian Wei
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiangyu Lu
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Qinghong Sun
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China
| | - Chuqiao Wang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jiuyan Zhang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Honggang Fan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
12
|
Wu L, Xiong X, Wu X, Ye Y, Jian Z, Zhi Z, Gu L. Targeting Oxidative Stress and Inflammation to Prevent Ischemia-Reperfusion Injury. Front Mol Neurosci 2020; 13:28. [PMID: 32194375 PMCID: PMC7066113 DOI: 10.3389/fnmol.2020.00028] [Citation(s) in RCA: 281] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
The cerebral ischemia injury can result in neuronal death and/or functional impairment, which leads to further damage and dysfunction after recovery of blood supply. Cerebral ischemia/reperfusion injury (CIRI) often causes irreversible brain damage and neuronal injury and death, which involves many complex pathological processes including oxidative stress, amino acid toxicity, the release of endogenous substances, inflammation and apoptosis. Oxidative stress and inflammation are interactive and play critical roles in ischemia/reperfusion injury in the brain. Oxidative stress is important in the pathological process of ischemic stroke and is critical for the cascade development of ischemic injury. Oxidative stress is caused by reactive oxygen species (ROS) during cerebral ischemia and is more likely to lead to cell death and ultimately brain death after reperfusion. During reperfusion especially, superoxide anion free radicals, hydroxyl free radicals, and nitric oxide (NO) are produced, which can cause lipid peroxidation, inflammation and cell apoptosis. Inflammation alters the balance between pro-inflammatory and anti-inflammatory factors in cerebral ischemic injury. Inflammatory factors can therefore stimulate or exacerbate inflammation and aggravate ischemic injury. Neuroprotective therapies for various stages of the cerebral ischemia cascade response have received widespread attention. At present, neuroprotective drugs mainly include free radical scavengers, anti-inflammatory agents, and anti-apoptotic agents. However, the molecular mechanisms of the interaction between oxidative stress and inflammation, and their interplay with different types of programmed cell death in ischemia/reperfusion injury are unclear. The development of a suitable method for combination therapy has become a hot topic.
Collapse
Affiliation(s)
- Liquan Wu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaomin Wu
- Department of Anesthesiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Yingze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zeng Zhi
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
13
|
Thome JG, Reeder EL, Collins SM, Gopalan P, Robson MJ. Contributions of Interleukin-1 Receptor Signaling in Traumatic Brain Injury. Front Behav Neurosci 2020; 13:287. [PMID: 32038189 PMCID: PMC6985078 DOI: 10.3389/fnbeh.2019.00287] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/16/2019] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) in various forms affects millions in the United States annually. There are currently no FDA-approved therapies for acute injury or the chronic comorbidities associated with TBI. Acute phases of TBI are characterized by profound neuroinflammation, a process that stimulates the generation and release of proinflammatory cytokines including interleukin-1α (IL-1α) and IL-1β. Both forms of IL-1 initiate signaling by binding with IL-1 receptor type 1 (IL-1R1), a receptor with a natural, endogenous antagonist dubbed IL-1 receptor antagonist (IL-1Ra). The recombinant form of IL-1Ra has gained FDA approval for inflammatory conditions such as rheumatoid arthritis, prompting interest in repurposing these pharmacotherapies for other inflammatory diseases/injury states including TBI. This review summarizes the currently available preclinical and clinical literature regarding the therapeutic potential of inhibiting IL-1-mediated signaling in the context of TBI. Additionally, we propose specific research areas that would provide a greater understanding of the role of IL-1 signaling in TBI and how these data may be beneficial for the development of IL-1-targeted therapies, ushering in the first FDA-approved pharmacotherapy for acute TBI.
Collapse
Affiliation(s)
- Jason G Thome
- Department of Anesthesia and Critical Care, Division of Biological Sciences, College of Medicine, University of Chicago, Chicago, IL, United States
| | - Evan L Reeder
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, United States
| | - Sean M Collins
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, United States
| | - Poornima Gopalan
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, United States
| | - Matthew J Robson
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
14
|
Lambertsen KL, Finsen B, Clausen BH. Post-stroke inflammation-target or tool for therapy? Acta Neuropathol 2019; 137:693-714. [PMID: 30483945 PMCID: PMC6482288 DOI: 10.1007/s00401-018-1930-z] [Citation(s) in RCA: 297] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/03/2018] [Accepted: 11/04/2018] [Indexed: 12/22/2022]
Abstract
Inflammation is currently considered a prime target for the development of new stroke therapies. In the acute phase of ischemic stroke, microglia are activated and then circulating immune cells invade the peri-infarct and infarct core. Resident and infiltrating cells together orchestrate the post-stroke inflammatory response, communicating with each other and the ischemic neurons, through soluble and membrane-bound signaling molecules, including cytokines. Inflammation can be both detrimental and beneficial at particular stages after a stroke. While it can contribute to expansion of the infarct, it is also responsible for infarct resolution, and influences remodeling and repair. Several pre-clinical and clinical proof-of-concept studies have suggested the effectiveness of pharmacological interventions that target inflammation post-stroke. Experimental evidence shows that targeting certain inflammatory cytokines, such as tumor necrosis factor, interleukin (IL)-1, IL-6, and IL-10, holds promise. However, as these cytokines possess non-redundant protective and immunoregulatory functions, their neutralization or augmentation carries a risk of unwanted side effects, and clinical translation is, therefore, challenging. This review summarizes the cell biology of the post-stroke inflammatory response and discusses pharmacological interventions targeting inflammation in the acute phase after a stroke that may be used alone or in combination with recanalization therapies. Development of next-generation immune therapies should ideally aim at selectively neutralizing pathogenic immune signaling, enhancing tissue preservation, promoting neurological recovery and leaving normal function intact.
Collapse
Affiliation(s)
- Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense, Denmark.
- Department of Clinical Research, BRIDGE-Brain Research-Inter-Disciplinary Guided Excellence, University of Southern Denmark, 5000, Odense C, Denmark.
- Department of Neurology, Odense University Hospital, 5000, Odense, Denmark.
| | - Bente Finsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense, Denmark
- Department of Clinical Research, BRIDGE-Brain Research-Inter-Disciplinary Guided Excellence, University of Southern Denmark, 5000, Odense C, Denmark
| | - Bettina Hjelm Clausen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense, Denmark
- Department of Clinical Research, BRIDGE-Brain Research-Inter-Disciplinary Guided Excellence, University of Southern Denmark, 5000, Odense C, Denmark
| |
Collapse
|
15
|
Acute transient cognitive dysfunction and acute brain injury induced by systemic inflammation occur by dissociable IL-1-dependent mechanisms. Mol Psychiatry 2019; 24:1533-1548. [PMID: 29875474 PMCID: PMC6510649 DOI: 10.1038/s41380-018-0075-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 02/12/2018] [Accepted: 04/03/2018] [Indexed: 01/06/2023]
Abstract
Systemic inflammation can impair cognition with relevance to dementia, delirium and post-operative cognitive dysfunction. Episodes of delirium also contribute to rates of long-term cognitive decline, implying that these acute events induce injury. Whether systemic inflammation-induced acute dysfunction and acute brain injury occur by overlapping or discrete mechanisms remains unexplored. Here we show that systemic inflammation, induced by bacterial LPS, produces both working-memory deficits and acute brain injury in the degenerating brain and that these occur by dissociable IL-1-dependent processes. In normal C57BL/6 mice, LPS (100 µg/kg) did not affect working memory but impaired long-term memory consolidation. However prior hippocampal synaptic loss left mice selectively vulnerable to LPS-induced working memory deficits. Systemically administered IL-1 receptor antagonist (IL-1RA) was protective against, and systemic IL-1β replicated, these working memory deficits. Dexamethasone abolished systemic cytokine synthesis and was protective against working memory deficits, without blocking brain IL-1β synthesis. Direct application of IL-1β to ex vivo hippocampal slices induced non-synaptic depolarisation and irreversible loss of membrane potential in CA1 neurons from diseased animals and systemic LPS increased apoptosis in the degenerating brain, in an IL-1RI-dependent fashion. The data suggest that LPS induces working memory dysfunction via circulating IL-1β but direct hippocampal action of IL-1β causes neuronal dysfunction and may drive neuronal death. The data suggest that acute systemic inflammation produces both reversible cognitive deficits, resembling delirium, and acute brain injury contributing to long-term cognitive impairment but that these events are mechanistically dissociable. These data have significant implications for management of cognitive dysfunction during acute illness.
Collapse
|
16
|
Abstract
BACKGROUND AND OBJECTIVE Cerebral ischemia affects hepatic enzymes and brain permeability extensively. Piracetam was investigated up to phase III of clinical trials and there is lack of data on brain penetration in cerebral ischemic condition. Thus, knowledge of the pharmacokinetics and brain penetration of piracetam during ischemic condition would aid to improve pharmacotherapeutics in ischemic stroke. METHODS Focal cerebral ischemia was induced by middle cerebral artery occlusion for 2 h in male Wistar rats followed by reperfusion. After 24 h of middle cerebral artery occlusion or 22 h of reperfusion, piracetam was administered for pharmacokinetic, brain penetration, and pharmacological experiments. In pharmacokinetic study, blood samples were collected at different time points after 200-mg/kg (oral) and 75-mg/kg (intravenous) administration of piracetam through right external jugular vein cannulation. In brain penetration study, the cerebrospinal fluid, systemic blood, portal blood, and brain samples were collected at pre-designated time points after 200-mg/kg oral administration of piracetam. In a separate experiment, the pharmacological effect of the single oral dose of piracetam in middle cerebral artery occlusion was assessed at a dose of 200 mg/kg. RESULTS All the pharmacokinetic parameters of piracetam including area under curve (AUC0-24), maximum plasma concentration (C max), time to reach the maximum plasma concentration (t max), elimination half-life (t 1/2), volume of distribution (V z), total body clearance, mean residence time, and bioavailability were found to be similar in ischemic stroke condition except for brain penetration. Piracetam exposure (AUC0-2) in brain and CSF were found to be 2.4- and 3.1-fold higher, respectively, in ischemic stroke compared to control rats. Piracetam significantly reduced infarct volume by 35.77% caused by middle cerebral artery occlusion. CONCLUSIONS There was no change in the pharmacokinetic parameters of piracetam in the ischemic stroke model except for brain penetration. This indicates that variables influencing brain penetration may not be limiting factors for use of piracetam in ischemic stroke.
Collapse
|
17
|
Combined Blockade of Interleukin-1α and -1β Signaling Protects Mice from Cognitive Dysfunction after Traumatic Brain Injury. eNeuro 2018; 5:eN-NWR-0385-17. [PMID: 29662944 PMCID: PMC5898697 DOI: 10.1523/eneuro.0385-17.2018] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/27/2018] [Accepted: 03/15/2018] [Indexed: 12/20/2022] Open
Abstract
Diffuse activation of interleukin-1 inflammatory cytokine signaling after traumatic brain injury (TBI) elicits progressive neurodegeneration and neuropsychiatric dysfunction, and thus represents a potential opportunity for therapeutic intervention. Although interleukin (IL)-1α and IL-1β both activate the common type 1 IL-1 receptor (IL-1RI), they manifest distinct injury-specific roles in some models of neurodegeneration. Despite its potential relevance to treating patients with TBI, however, the individual contributions of IL-1α and IL-1β to TBI-pathology have not been previously investigated. To address this need, we applied genetic and pharmacologic approaches in mice to dissect the individual contributions of IL-1α, IL-β, and IL-1RI signaling to the pathophysiology of fluid percussion–mediated TBI, a model of mixed focal and diffuse TBI. IL-1RI ablation conferred a greater protective effect on brain cytokine expression and cognitive function after TBI than did individual IL-1α or IL-1β ablation. This protective effect was recapitulated by treatment with the drug anakinra, a recombinant naturally occurring IL-1RI antagonist. Our data thus suggest that broad targeting of IL-1RI signaling is more likely to reduce neuroinflammation and preserve cognitive function after TBI than are approaches that individually target IL-1α or IL-1β signaling.
Collapse
|
18
|
Qi Y, Klyubin I, Cuello AC, Rowan MJ. NLRP3-dependent synaptic plasticity deficit in an Alzheimer's disease amyloidosis model in vivo. Neurobiol Dis 2018; 114:24-30. [PMID: 29477641 DOI: 10.1016/j.nbd.2018.02.016] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/24/2018] [Accepted: 02/21/2018] [Indexed: 01/12/2023] Open
Abstract
Pro-inflammatory mechanisms have recently emerged as an important component of early Alzheimer's disease (AD) pathogenesis. A particularly attractive therapeutic strategy is to selectively prevent the disruptive effects of activation of the innate immune system in the brain at an early transitional stage by reducing the production or directly neutralizing pro-inflammatory cytokines, in particular IL-1β and TNF-α. Here we tested their in vivo effects on synaptic plasticity deficits, which provide sensitive and robust measures of synaptic failure, in a rat model of AD amyloidosis. Using electrophysiological techniques we longitudinally studied the effects of the NLRP3 inflammasome inhibitor Mcc950, the IL-1 receptor antagonist (anakinra) and an anti-TNF-α agent (etanercept) in awake freely moving transgenic rats overexpressing AD associated β-amyloid precursor protein at a pre-plaque stage of amyloidosis. Repeated treatment with Mcc950 reversibly abrogated the inhibition of long-term potentiation. The IL-1 receptor antagonist and etanercept also had a similar beneficial effect on the deficit in synaptic plasticity. Our findings support the clinical development of Mcc950 and clinically available IL-1- and TNF-α-neutralizing agents in early AD.
Collapse
Affiliation(s)
- Yingjie Qi
- Department of Pharmacology and Therapeutics, Watts Building, Trinity College Dublin, Dublin 2, Ireland; Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Igor Klyubin
- Department of Pharmacology and Therapeutics, Watts Building, Trinity College Dublin, Dublin 2, Ireland; Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland.
| | - A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, 3655 Sir-William-Osler Promenade, Room 1210, Montreal, QC H3G1Y6, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal H3G1Y6, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal H3G1Y6, Canada
| | - Michael J Rowan
- Department of Pharmacology and Therapeutics, Watts Building, Trinity College Dublin, Dublin 2, Ireland; Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
19
|
Sun M, Brady RD, Wright DK, Kim HA, Zhang SR, Sobey CG, Johnstone MR, O'Brien TJ, Semple BD, McDonald SJ, Shultz SR. Treatment with an interleukin-1 receptor antagonist mitigates neuroinflammation and brain damage after polytrauma. Brain Behav Immun 2017; 66:359-371. [PMID: 28782716 DOI: 10.1016/j.bbi.2017.08.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 07/22/2017] [Accepted: 08/02/2017] [Indexed: 01/18/2023] Open
Abstract
Traumatic brain injury (TBI) and long bone fracture are common in polytrauma. This injury combination in mice results in elevated levels of the pro-inflammatory cytokine interleukin-1β (IL-1β) and exacerbated neuropathology when compared to isolated-TBI. Here we examined the effect of treatment with an IL-1 receptor antagonist (IL-1ra) in mice given a TBI and a concomitant tibial fracture (i.e., polytrauma). Adult male C57BL/6 mice were given sham-injuries or polytrauma and treated with saline-vehicle or IL-1ra (100mg/kg). Treatments were subcutaneously injected at 1, 6, and 24h, and then once daily for one week post-injury. 7-8 mice/group were euthanized at 48h post-injury. 12-16 mice/group underwent behavioral testing at 12weeks post-injury and MRI at 14weeks post-injury before being euthanized at 16weeks post-injury. At 48h post-injury, markers for activated microglia and astrocytes, as well as neutrophils and edema, were decreased in polytrauma mice treated with IL-1ra compared to polytrauma mice treated with vehicle. At 14weeks post-injury, MRI analysis demonstrated that IL-1ra treatment after polytrauma reduced volumetric loss in the injured cortex and mitigated track-weighted MRI markers for axonal injury. As IL-1ra (Anakinra) is approved for human use, it may represent a promising therapy in polytrauma cases involving TBI and fracture.
Collapse
Affiliation(s)
- Mujun Sun
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC 3052, Australia
| | - Rhys D Brady
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC 3052, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, VIC 3083, Australia
| | - David K Wright
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC 3052, Australia; The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia; Departments of Neuroscience and Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Hyun Ah Kim
- Department of Physiology, Anatomy and Microbiology, La Trobe University, VIC 3083, Australia
| | - Shenpeng R Zhang
- Department of Physiology, Anatomy and Microbiology, La Trobe University, VIC 3083, Australia; Department of Pharmacology, Monash University, Melbourne, VIC 3800, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, VIC 3083, Australia
| | - Maddison R Johnstone
- Department of Physiology, Anatomy and Microbiology, La Trobe University, VIC 3083, Australia
| | - Terence J O'Brien
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC 3052, Australia; Departments of Neuroscience and Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Bridgette D Semple
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC 3052, Australia; Departments of Neuroscience and Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Stuart J McDonald
- Department of Physiology, Anatomy and Microbiology, La Trobe University, VIC 3083, Australia
| | - Sandy R Shultz
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC 3052, Australia; Departments of Neuroscience and Medicine, Monash University, Melbourne, VIC 3004, Australia.
| |
Collapse
|
20
|
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
|
21
|
Clark IA, Vissel B. The meteorology of cytokine storms, and the clinical usefulness of this knowledge. Semin Immunopathol 2017; 39:505-516. [PMID: 28451786 PMCID: PMC5495849 DOI: 10.1007/s00281-017-0628-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 04/10/2017] [Indexed: 01/07/2023]
Abstract
The term cytokine storm has become a popular descriptor of the dramatic harmful consequences of the rapid release of polypeptide mediators, or cytokines, that generate inflammatory responses. This occurs throughout the body in both non-infectious and infectious disease states, including the central nervous system. In infectious disease it has become a useful concept through which to appreciate that most infectious disease is not caused directly by a pathogen, but by an overexuberant innate immune response by the host to its presence. It is less widely known that in addition to these roles in disease pathogenesis these same cytokines are also the basis of innate immunity, and in lower concentrations have many essential physiological roles. Here we update this field, including what can be learned through the history of how these interlinking three aspects of biology and disease came to be appreciated. We argue that understanding cytokine storms in their various degrees of acuteness, severity and persistence is essential in order to grasp the pathophysiology of many diseases, and thus the basis of newer therapeutic approaches to treating them. This particularly applies to the neurodegenerative diseases.
Collapse
Affiliation(s)
- Ian A Clark
- Research School of Biology, Australian National University, Canberra, Australia.
| | - Bryce Vissel
- School of Life Sciences, Faculty of Science, University of Technology, Sydney, Australia
- Garvan Institute of Medical Research, Sydney, Australia
| |
Collapse
|
22
|
Zhang DD, Zou MJ, Zhang YT, Fu WL, Xu T, Wang JX, Xia WR, Huang ZG, Gan XD, Zhu XM, Xu DG. A novel IL-1RA-PEP fusion protein with enhanced brain penetration ameliorates cerebral ischemia-reperfusion injury by inhibition of oxidative stress and neuroinflammation. Exp Neurol 2017; 297:1-13. [PMID: 28602833 DOI: 10.1016/j.expneurol.2017.06.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/14/2017] [Accepted: 06/07/2017] [Indexed: 12/21/2022]
Abstract
Neuroinflammation and oxidative stress are involved in cerebral ischemia-reperfusion, in which Interleukin 1 (IL-1), as an effective intervention target, is implicated. Interleukin-1 receptor antagonist (IL-1RA) is the natural inhibitor of IL-1, but blood-brain barrier (BBB) limits the brain penetration of intravenously administered IL-1RA, thereby restricting its therapeutic effect against neuroinflammation. In this study, we evaluated the potential effects of anti-inflammation and anti-oxidative stress of a novel protein IL-1RA-PEP, which fused IL-1RA with a cell penetrating peptide (CPP). Studies were carried out in transient middle cerebral artery occlusion (MCAO) in rats and oxygen glucose deprivation/reoxygenation (OGD/R) in primary cortical neurons. In MCAO rat model, IL-1RA-PEP (50mg/kg) injected i.v., penetrated BBB effectively, and alleviated brain infarction, cerebral edema, neurological deficit score and motor performance as well as inhibited the inflammatory cytokines expression. Furthermore, our results firstly showed that IL-1RA-PEP also regulated the oxidases expression, decreased the levels of NO, MDA and ROS. In addition, the inhibitory effects of IL-1RA-PEP on oxidative stress and inflammation were confirmed in rat cortical neurons induced by OGD/R, it reduced ROS, IL-6 and TNF-α. Further study showed that the effects of IL-1RA-PEP were closely associated with the NF-κB and p38 pathways which were proved respectively by their inhibitors JSH-23 and SB203580. Our results indicated that IL-1RA-PEP could effectively penetrate the brain of MCAO rats, alleviated the cerebral ischemia reperfusion injury by inhibiting neuroinflammation and oxidative stress, showing a great clinical potential for stroke.
Collapse
Affiliation(s)
- Dong-Dong Zhang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China; Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China
| | - Min-Ji Zou
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Ya-Tao Zhang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Wen-Liang Fu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Tao Xu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Jia-Xi Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Wen-Rong Xia
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Zhi-Guang Huang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Xiang-Dong Gan
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Xiao-Ming Zhu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China
| | - Dong-Gang Xu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, PR China; Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China.
| |
Collapse
|
23
|
Reparative effects of interleukin-1 receptor antagonist in young and aged/co-morbid rodents after cerebral ischemia. Brain Behav Immun 2017; 61:117-126. [PMID: 27856349 PMCID: PMC5325120 DOI: 10.1016/j.bbi.2016.11.013] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 10/21/2016] [Accepted: 11/12/2016] [Indexed: 11/21/2022] Open
Abstract
Neuroprotective strategies for ischemic stroke have failed to translate from bench to bedside, possibly due to the lack of consideration of key clinical co-morbidities. Stroke and co-morbidities are associated with raised levels of the pro-inflammatory cytokine interleukin-1 (IL-1). Inhibition of IL-1 by the administration of interleukin-1 receptor antagonist (IL-1Ra) has shown to be neuroprotective after experimental cerebral ischemia. Stroke can also trigger a robust neuroreparative response following injury, yet many of these new born neurons fail to survive or integrate into pre-existing circuits. Thus, we explore here effects of IL-1Ra on post-stroke neurogenesis in young and aged/co-morbid rats. Aged lean, aged Corpulent (a model of atherosclerosis, obesity and insulin resistance) and young Wistar male rats were exposed to transient cerebral ischemia, received subcutaneous IL-1Ra 3 and 6h during reperfusion, and effects on stroke outcome and neurogenesis were analyzed. Our results show that administration of IL-1Ra improves stroke outcome in both young and aged/co-morbid rats. Furthermore, IL-1Ra not only increases stem cell proliferation, but also significantly enhances neuroblast migration and the number of newly born neurons after cerebral ischemia. Overall, our data demonstrate that systemic administration of IL-1Ra improves outcome and promotes neurogenesis after experimental stroke, further highlighting the therapeutic potential of this clinically approved drug.
Collapse
|
24
|
Lee JH, Kam EH, Kim JM, Kim SY, Kim EJ, Cheon SY, Koo BN. Intranasal Administration of Interleukin-1 Receptor Antagonist in a Transient Focal Cerebral Ischemia Rat Model. Biomol Ther (Seoul) 2017; 25:149-157. [PMID: 27530114 PMCID: PMC5340539 DOI: 10.4062/biomolther.2016.050] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/06/2016] [Accepted: 05/24/2016] [Indexed: 11/05/2022] Open
Abstract
The interleukin-1 receptor antagonist (IL-1RA) is a potential stroke treatment candidate. Intranasal delivery is a novel method thereby a therapeutic protein can be penetrated into the brain parenchyma by bypassing the blood-brain barrier. Thus, this study tested whether intranasal IL-1RA can provide neuroprotection and brain penetration in transient cerebral ischemia. In male Sprague-Dawley rats, focal cerebral ischemia was induced by middle cerebral artery occlusion (MCAO) for 1 h. The rats simultaneously received 50 mg/kg human IL-1RA through the intranasal (IN group) or intraperitoneal route (IP group). The other rats were given 0.5 mL/kg normal saline (EC group). Neurobehavioral function, infarct size, and the concentration of the administered human IL-1RA in the brain tissue were assessed. In addition, the cellular distribution of intranasal IL-1RA in the brain and its effect on proinflammatory cytokines expression were evaluated. Intranasal IL-1RA improved neurological deficit and reduced infarct size until 7 days after MCAO (p<0.05). The concentrations of the human IL-1RA in the brain tissue 24 h after MCAO were significantly greater in the IN group than in the IP group (p<0.05). The human IL-1RA was confirmed to be co-localized with neuron and microglia. Furthermore, the IN group had lower expression of interleukin-1β and tumor necrosis factor-α at 6 h after MCAO than the EC group (p<0.05). These results suggest that intranasal IL-1RA can reach the brain parenchyma more efficiently and provide superior neuroprotection in the transient focal cerebral ischemia.
Collapse
Affiliation(s)
- Jae Hoon Lee
- Department of Anesthesiology and Pain Medicine, Severance Hospital, Seoul 03722, Republic of Korea.,Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Eun Hee Kam
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jeong Min Kim
- Department of Anesthesiology and Pain Medicine, Severance Hospital, Seoul 03722, Republic of Korea.,Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - So Yeon Kim
- Department of Anesthesiology and Pain Medicine, Severance Hospital, Seoul 03722, Republic of Korea.,Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Eun Jeong Kim
- Department of Anesthesiology and Pain Medicine, Severance Hospital, Seoul 03722, Republic of Korea.,Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - So Yeong Cheon
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Bon-Nyeo Koo
- Department of Anesthesiology and Pain Medicine, Severance Hospital, Seoul 03722, Republic of Korea.,Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
25
|
McCann SK, Cramond F, Macleod MR, Sena ES. Systematic Review and Meta-Analysis of the Efficacy of Interleukin-1 Receptor Antagonist in Animal Models of Stroke: an Update. Transl Stroke Res 2016; 7:395-406. [PMID: 27526101 PMCID: PMC5014900 DOI: 10.1007/s12975-016-0489-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 07/25/2016] [Accepted: 07/28/2016] [Indexed: 02/04/2023]
Abstract
Interleukin-1 receptor antagonist (IL-1 RA) is an anti-inflammatory protein used clinically to treat rheumatoid arthritis and is considered a promising candidate therapy for stroke. Here, we sought to update the existing systematic review and meta-analysis of IL-1 RA in models of ischaemic stroke, published in 2009, to assess efficacy, the range of circumstances in which efficacy has been tested and whether the data appear to be confounded due to reported study quality and publication bias. We included 25 sources of data, 11 of which were additional to the original review. Overall, IL-1 RA reduced infarct volume by 36.2 % (95 % confidence interval 31.6-40.7, n = 76 comparisons from 1283 animals). Assessments for publication bias suggest 30 theoretically missing studies which reduce efficacy to 21.9 % (17.3-26.4). Efficacy was higher where IL-1 RA was administered directly into the ventricles rather than peripherally, and studies not reporting allocation concealment during the induction of ischaemia reported larger treatment effects. The preclinical data supporting IL-1 RA as a candidate therapy for ischaemic stroke have improved. The reporting of measures to reduce the risk of bias has improved substantially in this update, and studies now include the use of animals with relevant co-morbidities.
Collapse
Affiliation(s)
- Sarah K McCann
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Fala Cramond
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Malcolm R Macleod
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Emily S Sena
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK.
| |
Collapse
|
26
|
Sobowale OA, Parry-Jones AR, Smith CJ, Tyrrell PJ, Rothwell NJ, Allan SM. Interleukin-1 in Stroke: From Bench to Bedside. Stroke 2016; 47:2160-7. [PMID: 26931154 DOI: 10.1161/strokeaha.115.010001] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 01/14/2016] [Indexed: 12/16/2022]
Affiliation(s)
- Oluwaseun A Sobowale
- From the Manchester Academic Health Sciences Centre, Salford Royal NHS Foundation Trust, University of Manchester, Salford, United Kingdom (O.A.S., A.R.P.-J., C.J.S., P.J.T.); Greater Manchester Neuroscience Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom (O.A.S., A.R.P.-J., C.J.S., P.J.T.); and Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom (N.J.R., S.M.A.)
| | - Adrian R Parry-Jones
- From the Manchester Academic Health Sciences Centre, Salford Royal NHS Foundation Trust, University of Manchester, Salford, United Kingdom (O.A.S., A.R.P.-J., C.J.S., P.J.T.); Greater Manchester Neuroscience Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom (O.A.S., A.R.P.-J., C.J.S., P.J.T.); and Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom (N.J.R., S.M.A.)
| | - Craig J Smith
- From the Manchester Academic Health Sciences Centre, Salford Royal NHS Foundation Trust, University of Manchester, Salford, United Kingdom (O.A.S., A.R.P.-J., C.J.S., P.J.T.); Greater Manchester Neuroscience Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom (O.A.S., A.R.P.-J., C.J.S., P.J.T.); and Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom (N.J.R., S.M.A.)
| | - Pippa J Tyrrell
- From the Manchester Academic Health Sciences Centre, Salford Royal NHS Foundation Trust, University of Manchester, Salford, United Kingdom (O.A.S., A.R.P.-J., C.J.S., P.J.T.); Greater Manchester Neuroscience Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom (O.A.S., A.R.P.-J., C.J.S., P.J.T.); and Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom (N.J.R., S.M.A.)
| | - Nancy J Rothwell
- From the Manchester Academic Health Sciences Centre, Salford Royal NHS Foundation Trust, University of Manchester, Salford, United Kingdom (O.A.S., A.R.P.-J., C.J.S., P.J.T.); Greater Manchester Neuroscience Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom (O.A.S., A.R.P.-J., C.J.S., P.J.T.); and Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom (N.J.R., S.M.A.)
| | - Stuart M Allan
- From the Manchester Academic Health Sciences Centre, Salford Royal NHS Foundation Trust, University of Manchester, Salford, United Kingdom (O.A.S., A.R.P.-J., C.J.S., P.J.T.); Greater Manchester Neuroscience Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom (O.A.S., A.R.P.-J., C.J.S., P.J.T.); and Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom (N.J.R., S.M.A.).
| |
Collapse
|
27
|
de Rivero Vaccari JP, Dietrich WD, Keane RW. Therapeutics targeting the inflammasome after central nervous system injury. Transl Res 2016; 167:35-45. [PMID: 26024799 PMCID: PMC4643411 DOI: 10.1016/j.trsl.2015.05.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 04/20/2015] [Accepted: 05/05/2015] [Indexed: 12/15/2022]
Abstract
Innate immunity is part of the early response of the body to deal with tissue damage and infections. Because of the early nature of the innate immune inflammatory response, this inflammatory reaction represents an attractive option as a therapeutic target. The inflammasome is a component of the innate immune response involved in the activation of caspase 1 and the processing of pro-interleukin 1β. In this article, we discuss the therapeutic potential of the inflammasome after central nervous system (CNS) injury and stroke, as well as the basic knowledge we have gained so far regarding inflammasome activation in the CNS. In addition, we discuss some of the therapies available or under investigation for the treatment of brain injury, spinal cord injury, and stroke.
Collapse
Affiliation(s)
- Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Fla.
| | - W Dalton Dietrich
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Fla
| | - Robert W Keane
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Fla; Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Fla
| |
Collapse
|
28
|
Therapies negating neuroinflammation after brain trauma. Brain Res 2015; 1640:36-56. [PMID: 26740405 DOI: 10.1016/j.brainres.2015.12.024] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 12/07/2015] [Accepted: 12/14/2015] [Indexed: 12/11/2022]
Abstract
Traumatic brain injury (TBI) elicits a complex secondary injury response, with neuroinflammation as a crucial central component. Long thought to be solely a deleterious factor, the neuroinflammatory response has recently been shown to be far more intricate, with both beneficial and detrimental consequences depending on the timing, magnitude and specific immune composition of the response post-injury. Despite extensive preclinical and clinical research into mechanisms of secondary injury after TBI, no effective neuroprotective therapy has been identified, with potential candidates repeatedly proving disappointing in the clinic. The neuroinflammatory response offers a promising avenue for therapeutic targeting, aiming to quell the deleterious consequences without influencing its function in providing a neurotrophic environment supportive of repair. The present review firstly describes the findings of recent clinical trials that aimed to modulate inflammation as a means of neuroprotection. Secondly, we discuss promising multifunctional and single-target anti-inflammatory candidates either currently in trial, or with ample experimental evidence supporting clinical application. This article is part of a Special Issue entitled SI:Brain injury and recovery.
Collapse
|
29
|
Nozu T, Kumei S, Miyagishi S, Takakusaki K, Okumura T. Colorectal distention induces acute and delayed visceral hypersensitivity: role of peripheral corticotropin-releasing factor and interleukin-1 in rats. J Gastroenterol 2015; 50:1153-61. [PMID: 25808230 DOI: 10.1007/s00535-015-1070-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/13/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Most studies evaluating visceral sensation measure visceromotor response (VMR) to colorectal distention (CRD). However, CRD itself induces visceral sensitization, and little is known about the detailed characteristics of this response. The present study tried to clarify this question. METHODS VMR was determined by measuring abdominal muscle contractions as a response to CRD in rats. The CRD set consisted of two isobaric distentions (60 mmHg for 10 min twice, with a 30-min rest), and the CRD set was performed on two separate days, i.e., days 1 and 3, 8. RESULTS On day 1, VMR to the second CRD was increased as compared with that to the first CRD, which is the acute sensitization. VMR to the first CRD on day 3 returned to the same level as that to the first CRD on day 1, and total VMR, i.e., the whole response to the CRD set, was not different between day 1 and day 3. However, total VMR was significantly increased on day 8 as compared with that on day 1, suggesting CRD induced the delayed sensitization. Intraperitoneally administered astressin (200 µg/kg), a corticotropin-releasing factor receptor antagonist, at the end of the first CRD blocked the acute sensitization, but anakinra (20 mg/kg, intraperitoneally), an interleukin-1 receptor antagonist, did not modify it. Astressin (200 µg/kg, twice before CRD on day 8) did not alter the delayed sensitization, but anakinra (20 mg/kg, twice) abolished it. CONCLUSIONS CRD induced both acute sensitization and delayed sensitization, which were mediated through peripheral corticotropin-releasing factor and interleukin-1 pathways, respectively.
Collapse
Affiliation(s)
- Tsukasa Nozu
- Department of Regional Medicine and Education, Asahikawa Medical University, Midorigaoka Higashi 2-1-1-1, Asahikawa, 078-8510, Japan.
| | - Shima Kumei
- Department of General Medicine, Asahikawa Medical University, Midorigaoka Higashi 2-1-1-1, Asahikawa, 078-8510, Japan
| | - Saori Miyagishi
- Department of General Medicine, Asahikawa Medical University, Midorigaoka Higashi 2-1-1-1, Asahikawa, 078-8510, Japan
| | - Kaoru Takakusaki
- Research Center for Brain Function and Medical Engineering, Asahikawa Medical University, Midorigaoka Higashi 2-1-1-1, Asahikawa, 078-8510, Japan
| | - Toshikatsu Okumura
- Department of General Medicine, Asahikawa Medical University, Midorigaoka Higashi 2-1-1-1, Asahikawa, 078-8510, Japan
| |
Collapse
|
30
|
Shevtsov MA, Nikolaev BP, Yakovleva LY, Dobrodumov AV, Zhakhov AV, Mikhrina AL, Pitkin E, Parr MA, Rolich VI, Simbircev AS, Ischenko AM. Recombinant interleukin-1 receptor antagonist conjugated to superparamagnetic iron oxide nanoparticles for theranostic targeting of experimental glioblastoma. Neoplasia 2015; 17:32-42. [PMID: 25622897 PMCID: PMC4309733 DOI: 10.1016/j.neo.2014.11.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 10/28/2014] [Accepted: 11/03/2014] [Indexed: 12/20/2022] Open
Abstract
Cerebral edema commonly accompanies brain tumors and contributes to neurologic symptoms. The role of the interleukin-1 receptor antagonist conjugated to superparamagnetic iron oxide nanoparticles (SPION-IL-1Ra) was assessed to analyze its anti-edemal effect and its possible application as a negative contrast enhancing agent for magnetic resonance imaging (MRI). Rats with intracranial C6 glioma were intravenously administered at various concentrations of IL-1Ra or SPION-IL-1Ra. Brain peritumoral edema following treatment with receptor antagonist was assessed with high-field MRI. IL-1Ra administered at later stages of tumor progression significantly reduced peritumoral edema (as measured by MRI) and prolonged two-fold the life span of comorbid animals in a dose-dependent manner in comparison to control and corticosteroid-treated animals (P < .001). Synthesized SPION-IL-1Ra conjugates had the properties of negative contrast agent with high coefficients of relaxation efficiency. In vitro studies of SPION-IL-1Ra nanoparticles demonstrated high intracellular incorporation and absence of toxic influence on C6 cells and lymphocyte viability and proliferation. Retention of the nanoparticles in the tumor resulted in enhanced hypotensive T2-weighted images of glioma, proving the application of the conjugates as negative magnetic resonance contrast agents. Moreover, nanoparticles reduced the peritumoral edema confirming the therapeutic potency of synthesized conjugates. SPION-IL-1Ra nanoparticles have an anti-edemal effect when administered through a clinically relevant route in animals with glioma. The SPION-IL-1Ra could be a candidate for theranostic approach in neuro-oncology both for diagnosis of brain tumors and management of peritumoral edema.
Collapse
Affiliation(s)
- Maxim A Shevtsov
- Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia; A.L. Polenov Russian Research Scientific Institute of Neurosurgery, St. Petersburg, Russia.
| | - Boris P Nikolaev
- Research Institute of Highly Pure Biopreparations, St. Petersburg, Russia
| | | | - Anatolii V Dobrodumov
- Institute of Macromolecular Compounds of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | | | - Anastasiy L Mikhrina
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Emil Pitkin
- Wharton School, University of Pennsylvania, Philadelphia, PA, USA
| | - Marina A Parr
- V.F. Fock Institute of Physics, St. Petersburg State University, St. Petersburg, Russia
| | - Valerii I Rolich
- V.F. Fock Institute of Physics, St. Petersburg State University, St. Petersburg, Russia
| | - Andrei S Simbircev
- Research Institute of Highly Pure Biopreparations, St. Petersburg, Russia
| | | |
Collapse
|
31
|
Brough D, Rothwell NJ, Allan SM. Interleukin-1 as a pharmacological target in acute brain injury. Exp Physiol 2015; 100:1488-94. [PMID: 26096539 DOI: 10.1113/ep085135] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 05/04/2015] [Accepted: 06/05/2015] [Indexed: 12/13/2022]
Abstract
NEW FINDINGS What is the topic of this review? This review discusses the latest findings on the contribution of inflammation to brain injury, how inflammation is a therapeutic target, and details of recent and forthcoming clinical studies. What advances does it highlight? Here we highlight recent advances on the role and regulation of inflammasomes, and the latest clinical progress in targeting inflammation. Acute brain injury is one of the leading causes of mortality and disability worldwide. Despite this, treatments for acute brain injuries are limited, and there remains a massive unmet clinical need. Inflammation has emerged as a major contributor to non-communicable diseases, and there is now substantial and growing evidence that inflammation, driven by the cytokine interleukin-1 (IL-1), worsens acute brain injury. Interleukin-1 is regulated by large, multimolecular complexes called inflammasomes. Here, we discuss the latest research on the regulation of inflammasomes and IL-1 in the brain, preclinical efforts to establish the IL-1 system as a therapeutic target, and the promise of recent and future clinical studies on blocking the action of IL-1 for the treatment of brain injury.
Collapse
Affiliation(s)
- David Brough
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Nancy J Rothwell
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Stuart M Allan
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
32
|
Xiong Y, Zhang Y, Mahmood A, Chopp M. Investigational agents for treatment of traumatic brain injury. Expert Opin Investig Drugs 2015; 24:743-60. [PMID: 25727893 PMCID: PMC4433440 DOI: 10.1517/13543784.2015.1021919] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Traumatic brain injury (TBI) is a major cause of death and disability worldwide. To date, there are no pharmacologic agents proven to improve outcomes from TBI because all the Phase III clinical trials in TBI have failed. Thus, there is a compelling need to develop treatments for TBI. AREAS COVERED The following article provides an overview of select cell-based and pharmacological therapies under early development for the treatment of TBI. These therapies seek to enhance cognitive and neurological functional recovery through neuroprotective and neurorestorative strategies. EXPERT OPINION TBI elicits both complex degenerative and regenerative tissue responses in the brain. TBI can lead to cognitive, behavioral, and motor deficits. Although numerous promising neuroprotective treatment options have emerged from preclinical studies that mainly target the lesion, translation of preclinical effective neuroprotective drugs to clinical trials has proven challenging. Accumulating evidence indicates that the mammalian brain has a significant, albeit limited, capacity for both structural and functional plasticity, as well as regeneration essential for spontaneous functional recovery after injury. A new therapeutic approach is to stimulate neurovascular remodeling by enhancing angiogenesis, neurogenesis, oligodendrogenesis, and axonal sprouting, which in concert, may improve neurological functional recovery after TBI.
Collapse
Affiliation(s)
- Ye Xiong
- Henry Ford Hospital, Department of Neurosurgery , Education and Research Building, Room 3096, 2799 West Grand Boulevard, Detroit, MI 48202 , USA +1 313 916 4743 ; +1 313 916 9855 ;
| | | | | | | |
Collapse
|
33
|
Smith CJ, Denes A, Tyrrell PJ, Di Napoli M. Phase II anti-inflammatory and immune-modulating drugs for acute ischaemic stroke. Expert Opin Investig Drugs 2015; 24:623-43. [PMID: 25727670 DOI: 10.1517/13543784.2015.1020110] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Stroke is the second leading cause of death worldwide and the leading cause of adult neurological disability. Despite advances in stroke unit care, and increasing use of thrombolysis, there remains an urgent need for safe and effective treatments for acute ischaemic stroke. However, this is against a backdrop of multiple failures in translational drug development. Cerebral ischaemia initiates a complex cascade of immune and inflammatory pathways in the brain microvasculature and periphery, which contribute to the evolution of cerebral injury, resolution and repair. Targeting specific inflammatory or immune pathways, therefore, represents an attractive treatment strategy in acute ischaemic stroke. Although anti-inflammatory drugs have already failed in clinical trial development, several are currently at the Phase II developmental stage. AREAS COVERED The authors highlight several candidate drugs, which modulate a range of inflammatory and immune pathways, and have been investigated in pre-clinical and Phase II studies to date. EXPERT OPINION Drugs targeting inflammatory and immune pathways offer theoretical advantages including potentially longer therapeutic time windows and effects complementary to thrombolysis (ameliorating reperfusion injury). Fundamental changes in the approach to pre-clinical and clinical drug development are required to facilitate successful translation of promising candidate drugs into clinical practice.
Collapse
Affiliation(s)
- Craig J Smith
- Greater Manchester Comprehensive Stroke Centre, Department of Medical Neurosciences, Salford Royal Foundation Trust , Salford , UK
| | | | | | | |
Collapse
|
34
|
Murray KN, Parry-Jones AR, Allan SM. Interleukin-1 and acute brain injury. Front Cell Neurosci 2015; 9:18. [PMID: 25705177 PMCID: PMC4319479 DOI: 10.3389/fncel.2015.00018] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 01/12/2015] [Indexed: 01/05/2023] Open
Abstract
Inflammation is the key host-defense response to infection and injury, yet also a major contributor to a diverse range of diseases, both peripheral and central in origin. Brain injury as a result of stroke or trauma is a leading cause of death and disability worldwide, yet there are no effective treatments, resulting in enormous social and economic costs. Increasing evidence, both preclinical and clinical, highlights inflammation as an important factor in stroke, both in determining outcome and as a contributor to risk. A number of inflammatory mediators have been proposed as key targets for intervention to reduce the burden of stroke, several reaching clinical trial, but as yet yielding no success. Many factors could explain these failures, including the lack of robust preclinical evidence and poorly designed clinical trials, in addition to the complex nature of the clinical condition. Lack of consideration in preclinical studies of associated co-morbidities prevalent in the clinical stroke population is now seen as an important omission in previous work. These co-morbidities (atherosclerosis, hypertension, diabetes, infection) have a strong inflammatory component, supporting the need for greater understanding of how inflammation contributes to acute brain injury. Interleukin (IL)-1 is the prototypical pro-inflammatory cytokine, first identified many years ago as the endogenous pyrogen. Research over the last 20 years or so reveals that IL-1 is an important mediator of neuronal injury and blocking the actions of IL-1 is beneficial in a number of experimental models of brain damage. Mechanisms underlying the actions of IL-1 in brain injury remain unclear, though increasing evidence indicates the cerebrovasculature as a key target. Recent literature supporting this and other aspects of how IL-1 and systemic inflammation in general contribute to acute brain injury are discussed in this review.
Collapse
Affiliation(s)
- Katie N Murray
- Faculty of Life Sciences, University of Manchester Manchester, UK
| | | | - Stuart M Allan
- Faculty of Life Sciences, University of Manchester Manchester, UK
| |
Collapse
|
35
|
Giles JA, Greenhalgh AD, Davies CL, Denes A, Shaw T, Coutts G, Rothwell NJ, McColl BW, Allan SM. Requirement for interleukin-1 to drive brain inflammation reveals tissue-specific mechanisms of innate immunity. Eur J Immunol 2014; 45:525-30. [PMID: 25367678 PMCID: PMC4357393 DOI: 10.1002/eji.201444748] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 09/30/2014] [Accepted: 10/22/2014] [Indexed: 01/08/2023]
Abstract
The immune system is implicated in a wide range of disorders affecting the brain and is, therefore, an attractive target for therapy. Interleukin-1 (IL-1) is a potent regulator of the innate immune system important for host defense but is also associated with injury and disease in the brain. Here, we show that IL-1 is a key mediator driving an innate immune response to inflammatory challenge in the mouse brain but is dispensable in extracerebral tissues including the lung and peritoneum. We also demonstrate that IL-1α is an important ligand contributing to the CNS dependence on IL-1 and that IL-1 derived from the CNS compartment (most likely microglia) is the major source driving this effect. These data reveal previously unknown tissue-specific requirements for IL-1 in driving innate immunity and suggest that IL-1-mediated inflammation in the brain could be selectively targeted without compromising systemic innate immune responses that are important for resistance to infection. This property could be exploited to mitigate injury- and disease-associated inflammation in the brain without increasing susceptibility to systemic infection, an important complication in several neurological disorders.
Collapse
Affiliation(s)
- James A Giles
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Codeine-induced hyperalgesia and allodynia: investigating the role of glial activation. Transl Psychiatry 2014; 4:e482. [PMID: 25386959 PMCID: PMC4259992 DOI: 10.1038/tp.2014.121] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 10/06/2014] [Indexed: 12/27/2022] Open
Abstract
Chronic morphine therapy has been associated with paradoxically increased pain. Codeine is a widely used opioid, which is metabolized to morphine to elicit analgesia. Prolonged morphine exposure exacerbates pain by activating the innate immune toll-like receptor-4 (TLR4) in the central nervous system. In silico docking simulations indicate codeine also docks to MD2, an accessory protein for TLR4, suggesting potential to induce TLR4-dependent pain facilitation. We hypothesized codeine would cause TLR4-dependent hyperalgesia/allodynia that is disparate from its opioid receptor-dependent analgesic rank potency. Hyperalgesia and allodynia were assessed using hotplate and von Frey tests at days 0, 3 and 5 in mice receiving intraperitoneal equimolar codeine (21 mg kg(-1)), morphine (20 mg kg(-1)) or saline, twice daily. This experiment was repeated in animals with prior partial nerve injury and in TLR4 null mutant mice. Interventions with interleukin-1 receptor antagonist (IL-1RA) and glial-attenuating drug ibudilast were assessed. Analyses of glial activation markers (glial fibrillary acid protein and CD11b) in neuronal tissue were conducted at the completion of behavioural testing. Despite providing less acute analgesia (P=0.006), codeine induced similar hotplate hyperalgesia to equimolar morphine vs saline (-9.5 s, P<0.01 and -7.3 s, P<0.01, respectively), suggesting codeine does not rely upon conversion to morphine to increase pain sensitivity. This highlights the potential non-opioid receptor-dependent nature of codeine-enhanced pain sensitivity-although the involvement of other codeine metabolites cannot be ruled out. IL-1RA reversed codeine-induced hyperalgesia (P<0.001) and allodynia (P<0.001), and TLR4 knock-out protected against codeine-induced changes in pain sensitivity. Glial attenuation with ibudilast reversed codeine-induced allodynia (P<0.001), and thus could be investigated further as potential treatment for codeine-induced pain enhancement.
Collapse
|
37
|
Bastien D, Lacroix S. Cytokine pathways regulating glial and leukocyte function after spinal cord and peripheral nerve injury. Exp Neurol 2014; 258:62-77. [PMID: 25017888 DOI: 10.1016/j.expneurol.2014.04.006] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Revised: 02/20/2014] [Accepted: 04/08/2014] [Indexed: 01/13/2023]
Abstract
Injury to the nervous system causes the almost immediate release of cytokines by glial cells and neurons. These cytokines orchestrate a complex array of responses leading to microgliosis, immune cell recruitment, astrogliosis, scarring, and the clearance of cellular debris, all steps that affect neuronal survival and repair. This review will focus on cytokines released after spinal cord and peripheral nerve injury and the primary signalling pathways triggered by these inflammatory mediators. Notably, the following cytokine families will be covered: IL-1, TNF, IL-6-like, TGF-β, and IL-10. Whether interfering with cytokine signalling could lead to novel therapies will also be discussed. Finally, the review will address whether manipulating the above-mentioned cytokine families and signalling pathways could exert distinct effects in the injured spinal cord versus peripheral nerve.
Collapse
Affiliation(s)
- Dominic Bastien
- Centre de recherche du Centre hospitalier universitaire de Québec-CHUL, Département de médecine moléculaire, Université Laval, Québec, QC, Canada
| | - Steve Lacroix
- Centre de recherche du Centre hospitalier universitaire de Québec-CHUL, Département de médecine moléculaire, Université Laval, Québec, QC, Canada..
| |
Collapse
|
38
|
Girard S, Murray KN, Rothwell NJ, Metz GAS, Allan SM. Long-term functional recovery and compensation after cerebral ischemia in rats. Behav Brain Res 2014; 270:18-28. [PMID: 24821402 PMCID: PMC4090421 DOI: 10.1016/j.bbr.2014.05.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 04/28/2014] [Accepted: 05/02/2014] [Indexed: 01/08/2023]
Abstract
Cerebral ischemia is one of the most common causes of disabilities in adults and leads to long-term motor and cognitive impairments with limited therapeutic possibilities. Treatment options have proven efficient in preclinical models of cerebral ischemia but have failed in the clinical setting. This limited translation may be due to the suitability of models used and outcomes measured as most studies have focused on the early period after injury with gross motor scales, which have limited correlation to the clinical situation. The aim of this study was to determine long-term functional outcomes after cerebral ischemia in rats, focusing on fine motor function, social and depressive behavior as clinically relevant measures. A secondary objective was to evaluate the effects of an anti-inflammatory treatment (interleukin-1 receptor antagonist (IL-1Ra)) on functional recovery and compensation. Infarct volume was correlated with long-term (25 days) impairments in fine motor skills, but not with emotional components of behavior. Motor impairments could not be detected using conventional neurological tests and only detailed analysis allowed differentiation between recovery and compensation. Acute systemic administration of IL-1Ra (at reperfusion) led to a faster and more complete recovery, but delayed (24h) IL-1Ra treatment had no effect. In summary functional assessment after brain injury requires detailed motor tests in order to address long-term impairments and compensation processes that are mediated by intact tissues. Functional deficits in skilled movement after brain injury represent ideal predictors of long-term outcomes and should become standard measures in the assessment of preclinical animal models.
Collapse
Affiliation(s)
- Sylvie Girard
- Faculty of Life Science, University of Manchester, Manchester, UK.
| | - Katie N Murray
- Faculty of Life Science, University of Manchester, Manchester, UK
| | - Nancy J Rothwell
- Faculty of Life Science, University of Manchester, Manchester, UK
| | - Gerlinde A S Metz
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Stuart M Allan
- Faculty of Life Science, University of Manchester, Manchester, UK
| |
Collapse
|
39
|
Amantea D, Certo M, Russo R, Bagetta G, Corasaniti MT, Tassorelli C. Early reperfusion injury is associated to MMP2 and IL-1β elevation in cortical neurons of rats subjected to middle cerebral artery occlusion. Neuroscience 2014; 277:755-63. [PMID: 25108165 DOI: 10.1016/j.neuroscience.2014.07.064] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 07/14/2014] [Accepted: 07/29/2014] [Indexed: 12/27/2022]
Abstract
The pathophysiological processes implicated in ischemic brain damage are strongly affected by an inflammatory reaction characterized by activation of immune cells and release of soluble mediators, including cytokines and chemokines. The pro-inflammatory cytokine interleukin (IL)-1β has been implicated in ischemic brain injury, however, to date, the mechanisms involved in the maturation of this cytokine in the ischemic brain have not been completely elucidated. We have previously suggested that matrix metalloproteinases (MMPs) may be implicated in cytokine production under pathological conditions. Here, we demonstrate that significant elevation of IL-1β occurs in the cortex as early as 1h after the beginning of reperfusion in rats subjected to 2-h middle cerebral artery occlusion (MCAo). At this early stage, we observe increased expression of IL-1β in pericallosal astroglial cells and in cortical neurons and this latter signal colocalizes with elevated gelatinolytic activity. By gel zymography, we demonstrate that the increased gelatinolytic signal at 1-h reperfusion is mainly ascribed to MMP2. Thus, MMP2 seems to contribute to early brain elevation of IL-β after transient ischemia and this mechanism may promote damage since pharmacological inhibition of gelatinases by the selective MMP2/MMP9 inhibitor V provides neuroprotection in rats subjected to transient MCAo.
Collapse
Affiliation(s)
- D Amantea
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Italy.
| | - M Certo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Italy
| | - R Russo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Italy
| | - G Bagetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Italy
| | - M T Corasaniti
- Department of Health Sciences, University Magna Graecia of Catanzaro, Italy
| | - C Tassorelli
- IRCCS National Neurological Institute C. Mondino Foundation, Pavia, Italy
| |
Collapse
|
40
|
Liu S, Zhu S, Zou Y, Wang T, Fu X. Knockdown of IL-1β improves hypoxia-ischemia brain associated with IL-6 up-regulation in cell and animal models. Mol Neurobiol 2014; 51:743-52. [PMID: 24965599 PMCID: PMC4359286 DOI: 10.1007/s12035-014-8764-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 05/26/2014] [Indexed: 10/25/2022]
Abstract
A study was conducted to investigate the effect of interleukin-1β (IL-1β) on hypoxia ischemia (HI) of cultured astrocyte and neonatal rat models and to explore the underlying molecular regulation mechanism. Primary rat astrocyte was exposed to hypoxia (2 % O2, 98 % N2) and cultured in serum-free medium for 6, 12, and 18 h to establish cell model of HI. Morphologic changes of astrocyte were monitored and gene expression change of IL-1β evaluated by real-time polymerase chain reaction (PCR). To establish the HI animal model, 3 days old postnatal Sprague-Dawley (SD) rats were treated with the right carotid artery ligation and were exposed to 8 % oxygen for 8, 16 and 24 h, respectively. Longa score scale, hematoxylin and eosin (HE) staining and water content were examined to assess neurologic function and morphology changes. IL-1β siRNA lentivirus (IL-1β-RNAi-LV) was injected into cerebral cortex motor area 2 days before HI and the interference efficiency examined by real-time PCR and Western blotting, respectively. Immunofluorescence staining of GFAP and IL-1β was performed to identify the location and interference effect of IL-1β, respectively. To further explore the potential mechanisms, the expression of inflammatory factors, including IL-6, IL-10 and tumor necrosis factor-alpha (TNF-α), was examined following IL-1β down-regulation. The size of soma astrocyte was increased greatly after 12 and 18 h of HI with IL-1β up-regulation. IL-1β knockdown by siRNA in vitro or by lentivirus in vivo can reverse cell swelling, brain edema and neurologic function deficiencies induced by HI. Lastly, interference of IL-1β remarkably increased IL-6 expression but not IL-10 and TNF-α. Therefore, down-regulation of IL-1β improves the deficiencies of neurologic function and morphology induced by HI, maybe closely associating with IL-6 regulation.
Collapse
Affiliation(s)
- Sujuan Liu
- Shenzhen Children's Hospital, Shenzhen, Guangdong, 518038, China
| | | | | | | | | |
Collapse
|
41
|
Shevtsov MA, Nikolaev BP, Yakovleva LY, Dobrodumov AV, Dayneko AS, Shmonin AA, Vlasov TD, Melnikova EV, Vilisov AD, Guzhova IV, Ischenko AM, Mikhrina AL, Galibin OV, Yakovenko IV, Margulis BA. Neurotherapeutic activity of the recombinant heat shock protein Hsp70 in a model of focal cerebral ischemia in rats. DRUG DESIGN DEVELOPMENT AND THERAPY 2014; 8:639-50. [PMID: 24920887 PMCID: PMC4044995 DOI: 10.2147/dddt.s62024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Recombinant 70 kDa heat shock protein (Hsp70) is an antiapoptotic protein that has a cell protective activity in stress stimuli and thus could be a useful therapeutic agent in the management of patients with acute ischemic stroke. The neuroprotective and neurotherapeutic activity of recombinant Hsp70 was explored in a model of experimental stroke in rats. Ischemia was produced by the occlusion of the middle cerebral artery for 45 minutes. To assess its neuroprotective capacity, Hsp70, at various concentrations, was intravenously injected 20 minutes prior to ischemia. Forty-eight hours after ischemia, rats were sacrificed and brain tissue sections were stained with 2% triphenyl tetrazolium chloride. Preliminary treatment with Hsp70 significantly reduced the ischemic zone (optimal response at 2.5 mg/kg). To assess Hsp70’s neurotherapeutic activity, we intravenously administered Hsp70 via the tail vein 2 hours after reperfusion (2 hours and 45 minutes after ischemia). Rats were then kept alive for 72 hours. The ischemic region was analyzed using a high-field 11 T MRI scanner. Administration of the Hsp70 decreased the infarction zone in a dose-dependent manner with an optimal (threefold) therapeutic response at 5 mg/kg. Long-term treatment of the ischemic rats with Hsp70 formulated in alginate granules with retarded release of protein further reduced the infarct volume in the brain as well as apoptotic area (annexin V staining). Due to its high neurotherapeutic potential, prolonged delivery of Hsp70 could be useful in the management of acute ischemic stroke.
Collapse
Affiliation(s)
- Maxim A Shevtsov
- Institute of Cytology of the Russian Academy of Sciences (RAS), St Petersburg, Russia ; AL Polenov Russian Research Scientific Institute of Neurosurgery, St Petersburg, Russia
| | - Boris P Nikolaev
- Research Institute of Highly Pure Biopreparations, St Petersburg, Russia
| | | | - Anatolii V Dobrodumov
- Institute of Macromolecular Compounds of the Russian Academy of Sciences (RAS), St Petersburg, Russia
| | - Anastasiy S Dayneko
- First St Petersburg IP Pavlov State Medical University, St Petersburg, Russia
| | - Alexey A Shmonin
- First St Petersburg IP Pavlov State Medical University, St Petersburg, Russia ; Federal Almazov Medical Research Centre, St Petersburg, Russia
| | - Timur D Vlasov
- First St Petersburg IP Pavlov State Medical University, St Petersburg, Russia
| | - Elena V Melnikova
- First St Petersburg IP Pavlov State Medical University, St Petersburg, Russia
| | - Alexander D Vilisov
- Institute of Macromolecular Compounds of the Russian Academy of Sciences (RAS), St Petersburg, Russia ; First St Petersburg IP Pavlov State Medical University, St Petersburg, Russia
| | - Irina V Guzhova
- Institute of Cytology of the Russian Academy of Sciences (RAS), St Petersburg, Russia
| | | | - Anastasiya L Mikhrina
- IM Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences (RAS), St Petersburg, Russia
| | - Oleg V Galibin
- First St Petersburg IP Pavlov State Medical University, St Petersburg, Russia
| | - Igor V Yakovenko
- AL Polenov Russian Research Scientific Institute of Neurosurgery, St Petersburg, Russia
| | - Boris A Margulis
- Institute of Cytology of the Russian Academy of Sciences (RAS), St Petersburg, Russia
| |
Collapse
|
42
|
Singh N, Hopkins SJ, Hulme S, Galea JP, Hoadley M, Vail A, Hutchinson PJ, Grainger S, Rothwell NJ, King AT, Tyrrell PJ. The effect of intravenous interleukin-1 receptor antagonist on inflammatory mediators in cerebrospinal fluid after subarachnoid haemorrhage: a phase II randomised controlled trial. J Neuroinflammation 2014; 11:1. [PMID: 24383930 PMCID: PMC3892121 DOI: 10.1186/1742-2094-11-1] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 12/17/2013] [Indexed: 12/12/2022] Open
Abstract
Background Interleukin-1 (IL-1) is a key mediator of ischaemic brain injury induced by stroke and subarachnoid haemorrhage (SAH). IL-1 receptor antagonist (IL-1Ra) limits brain injury in experimental stroke and reduces plasma inflammatory mediators associated with poor outcome in ischaemic stroke patients. Intravenous (IV) IL-1Ra crosses the blood–brain barrier (BBB) in patients with SAH, to achieve cerebrospinal fluid (CSF) concentrations that are neuroprotective in rats. Methods A small phase II, double-blind, randomised controlled study was carried out across two UK neurosurgical centres with the aim of recruiting 32 patients. Adult patients with aneurysmal SAH, requiring external ventricular drainage (EVD) within 72 hours of ictus, were eligible. Patients were randomised to receive IL-1Ra (500 mg bolus, then a 10 mg/kg/hr infusion for 24 hours) or placebo. Serial samples of CSF and plasma were taken and analysed for inflammatory mediators, with change in CSF IL-6 between 6 and 24 hours as the primary outcome measure. Results Six patients received IL-1Ra and seven received placebo. Concentrations of IL-6 in CSF and plasma were reduced by one standard deviation in the IL-1Ra group compared to the placebo group, between 6 and 24 hours, as predicted by the power calculation. This did not reach statistical significance (P = 0.08 and P = 0.06, respectively), since recruitment did not reach the target figure of 32. No adverse or serious adverse events reported were attributable to IL-1Ra. Conclusions IL-1Ra appears safe in SAH patients. The concentration of IL-6 was lowered to the degree expected, in both CSF and plasma for patients treated with IL-1Ra.
Collapse
Affiliation(s)
| | - Stephen J Hopkins
- The University of Manchester Stroke and Vascular Centre, Manchester Academic Health Sciences Centre, Clinical Sciences Building, Salford Royal NHS Foundation Trust, Stott Lane, Salford M6 8HD, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Rosenzweig JM, Lei J, Burd I. Interleukin-1 receptor blockade in perinatal brain injury. Front Pediatr 2014; 2:108. [PMID: 25340046 PMCID: PMC4187538 DOI: 10.3389/fped.2014.00108] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 09/23/2014] [Indexed: 12/13/2022] Open
Abstract
Interleukin-1 (IL-1) is a potent inflammatory cytokine that can be produced by a variety of cell types throughout the body. While IL-1 is a central mediator of inflammation and response to infection, the role of IL-1 signaling in adult and pediatric brain injury is becoming increasingly clear. Although the mechanisms of IL-1 expression are largely understood, the downstream effects and contributions to excitotoxicity and oxidative stress are poorly defined. Here, we present a review of mechanisms of IL-1 signaling with a focus on the role of IL-1 in perinatal brain injury. We highlight research models of perinatal brain injury and the use of interleukin-1 receptor antagonist (IL-1RA) as an agent of therapeutic potential in preventing perinatal brain injury due to exposure to inflammation.
Collapse
Affiliation(s)
- Jason M Rosenzweig
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Jun Lei
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Irina Burd
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine , Baltimore, MD , USA ; Department of Neuroscience, Kennedy Krieger Institute , Baltimore, MD , USA ; Department of Neurology, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| |
Collapse
|
44
|
Fann DYW, Lee SY, Manzanero S, Chunduri P, Sobey CG, Arumugam TV. Pathogenesis of acute stroke and the role of inflammasomes. Ageing Res Rev 2013; 12:941-66. [PMID: 24103368 DOI: 10.1016/j.arr.2013.09.004] [Citation(s) in RCA: 262] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Revised: 09/12/2013] [Accepted: 09/19/2013] [Indexed: 12/20/2022]
Abstract
Inflammation is an innate immune response to infection or tissue damage that is designed to limit harm to the host, but contributes significantly to ischemic brain injury following stroke. The inflammatory response is initiated by the detection of acute damage via extracellular and intracellular pattern recognition receptors, which respond to conserved microbial structures, termed pathogen-associated molecular patterns or host-derived danger signals termed damage-associated molecular patterns. Multi-protein complexes known as inflammasomes (e.g. containing NLRP1, NLRP2, NLRP3, NLRP6, NLRP7, NLRP12, NLRC4, AIM2 and/or Pyrin), then process these signals to trigger an effector response. Briefly, signaling through NLRP1 and NLRP3 inflammasomes produces cleaved caspase-1, which cleaves both pro-IL-1β and pro-IL-18 into their biologically active mature pro-inflammatory cytokines that are released into the extracellular environment. This review will describe the molecular structure, cellular signaling pathways and current evidence for inflammasome activation following cerebral ischemia, and the potential for future treatments for stroke that may involve targeting inflammasome formation or its products in the ischemic brain.
Collapse
|
45
|
Smith CJ, Lawrence CB, Rodriguez-Grande B, Kovacs KJ, Pradillo JM, Denes A. The immune system in stroke: clinical challenges and their translation to experimental research. J Neuroimmune Pharmacol 2013; 8:867-87. [PMID: 23673977 DOI: 10.1007/s11481-013-9469-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 04/28/2013] [Indexed: 12/27/2022]
Abstract
Stroke represents an unresolved challenge for both developed and developing countries and has a huge socio-economic impact. Although considerable effort has been made to limit stroke incidence and improve outcome, strategies aimed at protecting injured neurons in the brain have all failed. This failure is likely to be due to both the incompleteness of modelling the disease and its causes in experimental research, and also the lack of understanding of how systemic mechanisms lead to an acute cerebrovascular event or contribute to outcome. Inflammation has been implicated in all forms of brain injury and it is now clear that immune mechanisms profoundly influence (and are responsible for the development of) risk and causation of stroke, and the outcome following the onset of cerebral ischemia. Until very recently, systemic inflammatory mechanisms, with respect to common comorbidities in stroke, have largely been ignored in experimental studies. The main aim is therefore to understand interactions between the immune system and brain injury in order to develop novel therapeutic approaches. Recent data from clinical and experimental research clearly show that systemic inflammatory diseases -such as atherosclerosis, obesity, diabetes or infection - similar to stress and advanced age, are associated with dysregulated immune responses which can profoundly contribute to cerebrovascular inflammation and injury in the central nervous system. In this review, we summarize recent advances in the field of inflammation and stroke, focusing on the challenges of translation between pre-clinical and clinical studies, and potential anti-inflammatory/immunomodulatory therapeutic approaches.
Collapse
Affiliation(s)
- Craig J Smith
- Stroke and Vascular Research Centre, Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Science Centre, Salford Royal Foundation Trust, Salford M6 8HD, UK.
| | | | | | | | | | | |
Collapse
|
46
|
Bonestroo HJC, Nijboer CHA, van Velthoven CTJ, Kavelaars A, Hack CE, van Bel F, Heijnen CJ. Cerebral and hepatic inflammatory response after neonatal hypoxia-ischemia in newborn rats. Dev Neurosci 2013; 35:197-211. [PMID: 23689428 DOI: 10.1159/000346685] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 12/19/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Neonatal encephalopathy induced by perinatal asphyxia is a serious condition associated with high mortality and morbidity. Inflammation after the insult is thought to contribute to brain injury. This inflammatory response to hypoxia-ischemia (HI) may not only occur in the brain but also in peripheral organs. The aim of the present study was to investigate the effect of neonatal HI on the inflammatory response in the liver in comparison to inflammation in the brain. METHODS HI was induced in P7 Wistar rats by unilateral carotid artery occlusion and hypoxia. Cytokine and chemokine mRNA levels were determined in the brain and liver by quantitative PCR. Polarization of brain macrophages to the M1/M2-like phenotype and infiltration of neutrophils were characterized by immunohistochemistry. RESULTS 3 h after HI, an upregulation of the proinflammatory cytokines TNF-α and IL-1β and anti-inflammatory IL-10 was observed in the ipsilateral hemisphere of the brain compared to mRNA levels in sham-operated animals. Additionally, cerebral CINC-1 and MCP-1 mRNA expressions were increased. We also observed increased numbers of macrophages/microglia of the M1-like phenotype as well as a small increase in granulocyte influx in the ipsilateral hemisphere. Conversely, in the liver 3 h after HI, a downregulation of TNF-α, IL-1β, and MCP-1 and a trend towards an upregulation of IL-10 were observed compared to mRNA levels of sham-operated animals. However, hepatic CINC-1 expression was increased compared to levels in sham-operated animals. Following systemic hypoxia only, no significant changes in the expression of TNF-α, CINC-1 or MCP-1 were observed in the liver compared to sham-operated littermates, except for an upregulation in hepatic IL-1β expression 3 h after hypoxia. Twenty-four hours after insult, cerebral ipsilateral TNF-α, MCP-1 and CINC-1 mRNA expression was still increased, together with an increase in TGF-β expression. Moreover, an increase in macrophages/microglia of the M1-like phenotype was observed together with the appearance of macrophages/microglia of the M2-like phenotype around the cerebral lesion as well as an increase in granulocyte influx in comparison to 3 h after HI. In the liver, 24 h after HI, cytokine and chemokine responses were similar to mRNA levels in sham-operated animals except for a decrease in IL-10 and MCP-1. CONCLUSION We describe for the first time that brain damage following neonatal HI induces an early downregulation of the proinflammatory response in the liver. HI induces an early proinflammatory response in the brain with a concomitant increase in influx of neutrophils and polarization of macrophages/microglia to the M1-like phenotype starting at 3 h and increasing up to 24 h after HI. The inflammatory state of the brain changes after 24 h, with an increase in the anti-inflammatory cytokine TGF-β together with the appearance of macrophages/microglia of the M2-like phenotype. The downregulation of proinflammatory cytokines in the liver is not due to systemic hypoxia only, but is induced by the cerebral damage.
Collapse
Affiliation(s)
- H J C Bonestroo
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
47
|
Ström JO, Ingberg E, Theodorsson A, Theodorsson E. Method parameters' impact on mortality and variability in rat stroke experiments: a meta-analysis. BMC Neurosci 2013; 14:41. [PMID: 23548160 PMCID: PMC3637133 DOI: 10.1186/1471-2202-14-41] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 03/22/2013] [Indexed: 12/14/2022] Open
Abstract
Background Even though more than 600 stroke treatments have been shown effective in preclinical studies, clinically proven treatment alternatives for cerebral infarction remain scarce. Amongst the reasons for the discrepancy may be methodological shortcomings, such as high mortality and outcome variability, in the preclinical studies. A common approach in animal stroke experiments is that A) focal cerebral ischemia is inflicted, B) some type of treatment is administered and C) the infarct sizes are assessed. However, within this paradigm, the researcher has to make numerous methodological decisions, including choosing rat strain and type of surgical procedure. Even though a few studies have attempted to address the questions experimentally, a lack of consensus regarding the optimal methodology remains. Methods We therefore meta-analyzed data from 502 control groups described in 346 articles to find out how rat strain, procedure for causing focal cerebral ischemia and the type of filament coating affected mortality and infarct size variability. Results The Wistar strain and intraluminal filament procedure using a silicone coated filament was found optimal in lowering infarct size variability. The direct and endothelin methods rendered lower mortality rate, whereas the embolus method increased it compared to the filament method. Conclusions The current article provides means for researchers to adjust their middle cerebral artery occlusion (MCAo) protocols to minimize infarct size variability and mortality.
Collapse
Affiliation(s)
- Jakob O Ström
- Department of Clinical and Experimental Medicine, Clinical Chemistry, Faculty of Health Sciences, Linköping University, County Council of Östergötland, Linköping, Sweden.
| | | | | | | |
Collapse
|
48
|
Denes A, Wilkinson F, Bigger B, Chu M, Rothwell NJ, Allan SM. Central and haematopoietic interleukin-1 both contribute to ischaemic brain injury in mice. Dis Model Mech 2013; 6:1043-8. [PMID: 23519030 PMCID: PMC3701223 DOI: 10.1242/dmm.011601] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Interleukin-1 (IL-1) is a key regulator of inflammation and ischaemic brain injury, but the contribution of central and peripheral sources of IL-1 to brain injury is not well understood. Here we show that haematopoietic-derived IL-1 is a key driver of ischaemic brain injury. Wild type (WT) mice transplanted with IL-1αβ-deficient bone marrow displayed a significant (40%) reduction in brain injury induced by focal cerebral ischaemia compared with WT mice transplanted with WT bone marrow. This was paralleled by improved neurological outcome and the almost complete absence of splenic-derived, but not liver-derived, IL-1α after stroke in WT mice lacking haematopoietic-derived IL-1. IL-1αβ knockout (KO) mice transplanted with IL-1αβ-deficient bone marrow showed a 60% reduction in brain injury compared with WT mice receiving WT bone marrow. Transplantation of WT bone marrow in IL-1αβ KO mice resulted in a similar level of blood-brain-barrier injury to that observed in WT mice receiving IL-1αβ-deficient bone marrow. Cerebral oedema after brain injury was reduced in IL-1αβ KO recipients irrespective of donor-derived IL-1, but a lack of haematopoetic IL-1 has also been associated with smaller brain oedema independently of recipient status. Thus, both central and haematopoietic-derived IL-1 are important contributors to brain injury after cerebral ischaemia. Identification of the cellular sources of IL-1 in the periphery could allow targeted interventions at these sites.
Collapse
Affiliation(s)
- Adam Denes
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, UK
| | | | | | | | | | | |
Collapse
|
49
|
Kwon YS, Pineda E, Auvin S, Shin D, Mazarati A, Sankar R. Neuroprotective and antiepileptogenic effects of combination of anti-inflammatory drugs in the immature brain. J Neuroinflammation 2013; 10:30. [PMID: 23442201 PMCID: PMC3599749 DOI: 10.1186/1742-2094-10-30] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 02/15/2013] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Inflammatory signaling elicited by prolonged seizures can be contributory to neuronal injury as well as adverse plasticity leading to the development of spontaneous recurrent seizures (epilepsy) and associated co-morbidities. In this study, developing rat pups were subjected to lithium-pilocarpine status epilepticus (SE) at 2 and 3 weeks of age to study the effect of anti-inflammatory drugs (AID) on SE-induced hippocampal injury and the development of spontaneous seizures. FINDINGS We selected AIDs directed against interleukin-1 receptors (IL-1ra), a cyclooxygenase-2 (COX-2) inhibitor (CAY 10404), and an antagonist of microglia activation of caspase-1 (minocycline). Acute injury after SE was studied in the 2-week-old rats 24 h after SE. Development of recurrent spontaneous seizures was studied in 3-week-old rats subjected to SE 4 months after the initial insult.None of those AIDs were effective in attenuating CA1 injury in the 2-week-old pups or in limiting the development of spontaneous seizures in 3-week-old pups when administered individually. When empiric binary combinations of these drugs were tried, the combined targeting of IL-1r and COX-2 resulted in attenuation of acute CA1 injury, as determined 24 h after SE, in those animals. The same combination administered for 10 days following SE in 3-week-old rats, reduced the development of spontaneous recurrent seizures and limited the extent of mossy fiber sprouting. CONCLUSIONS Deployment of an empirically designed 'drug cocktail' targeting multiple inflammatory signaling pathways for a limited duration after an initial insult like SE may provide a practical approach to neuroprotection and anti-epileptogenic therapy.
Collapse
Affiliation(s)
- Young Se Kwon
- Department of Pediatrics, Division of Neurology, David Geffen School of Medicine at UCLA, 22-474 MDCC in CHS, Los Angeles, CA 90095-1752, USA
- Department of Pediatrics, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Eduardo Pineda
- Department of Pediatrics, Division of Neurology, David Geffen School of Medicine at UCLA, 22-474 MDCC in CHS, Los Angeles, CA 90095-1752, USA
| | - Stéphane Auvin
- Department of Pediatrics, Division of Neurology, David Geffen School of Medicine at UCLA, 22-474 MDCC in CHS, Los Angeles, CA 90095-1752, USA
- Department of Pediatric Neurology, Hôpital Robert Debré, INSERM U676, Paris, 75019, France
| | - Don Shin
- Department of Pediatrics, Division of Neurology, David Geffen School of Medicine at UCLA, 22-474 MDCC in CHS, Los Angeles, CA 90095-1752, USA
| | - Andrey Mazarati
- Department of Pediatrics, Division of Neurology, David Geffen School of Medicine at UCLA, 22-474 MDCC in CHS, Los Angeles, CA 90095-1752, USA
| | - Raman Sankar
- Department of Pediatrics, Division of Neurology, David Geffen School of Medicine at UCLA, 22-474 MDCC in CHS, Los Angeles, CA 90095-1752, USA
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
50
|
Influence of inflammation on poststroke plasticity. Neural Plast 2013; 2013:258582. [PMID: 23533818 PMCID: PMC3595668 DOI: 10.1155/2013/258582] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 01/11/2013] [Indexed: 01/17/2023] Open
Abstract
Age-related brain injuries including stroke are a leading cause of morbidity and mental disability worldwide. Most patients who survive stroke experience some degree of recovery. The restoration of lost functions can be explained by neuronal plasticity, understood as brain ability to reorganize and remodel itself in response to changed environmental requirements. However, stroke triggers a cascade of events which may prevent the normal development of the plastic changes. One of them may be inflammatory response initiated immediately after stroke, which has been found to contribute to neuronal injury. Some recent evidence though has suggested that inflammatory reaction can be also neuroprotective. This paper attempts to discuss the influence of poststroke inflammatory response on brain plasticity and stroke outcome. We also describe the recent anti-inflammatory strategies that have been effective for recovery in experimental stroke.
Collapse
|