1
|
Woodburn SC, Bollinger JL, Wohleb ES. The semantics of microglia activation: neuroinflammation, homeostasis, and stress. J Neuroinflammation 2021; 18:258. [PMID: 34742308 PMCID: PMC8571840 DOI: 10.1186/s12974-021-02309-6] [Citation(s) in RCA: 366] [Impact Index Per Article: 91.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023] Open
Abstract
Microglia are emerging as critical regulators of neuronal function and behavior in nearly every area of neuroscience. Initial reports focused on classical immune functions of microglia in pathological contexts, however, immunological concepts from these studies have been applied to describe neuro-immune interactions in the absence of disease, injury, or infection. Indeed, terms such as 'microglia activation' or 'neuroinflammation' are used ubiquitously to describe changes in neuro-immune function in disparate contexts; particularly in stress research, where these terms prompt undue comparisons to pathological conditions. This creates a barrier for investigators new to neuro-immunology and ultimately hinders our understanding of stress effects on microglia. As more studies seek to understand the role of microglia in neurobiology and behavior, it is increasingly important to develop standard methods to study and define microglial phenotype and function. In this review, we summarize primary research on the role of microglia in pathological and physiological contexts. Further, we propose a framework to better describe changes in microglia1 phenotype and function in chronic stress. This approach will enable more precise characterization of microglia in different contexts, which should facilitate development of microglia-directed therapeutics in psychiatric and neurological disease.
Collapse
Affiliation(s)
- Samuel C Woodburn
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Justin L Bollinger
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
2
|
Estes ML, McAllister AK. Alterations in immune cells and mediators in the brain: it's not always neuroinflammation! Brain Pathol 2015; 24:623-30. [PMID: 25345893 DOI: 10.1111/bpa.12198] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 08/11/2014] [Indexed: 01/02/2023] Open
Abstract
Neuroinflammation was once a clearly defined term denoting pathological immune processes within the central nervous system (CNS). Historically, this term was used to indicate the four hallmarks of peripheral inflammaton that occur following severe CNS injuries, such as stroke, injury or infection. Recently, however, the definition of neuroinflammation has relaxed to the point that it is often now assumed to be present when even only a single classical hallmark of inflammation is measured. As a result, a wide range of disorders, from psychiatric to degenerative diseases, are now assumed to have an integral inflammatory component. Ironically, at the same time, research has revealed unexpected nonclassical immune actions of immune mediators and cells in the CNS in the absence of pathology, increasing the likelihood that homeostatic and adaptive immune processes in the CNS will be mistaken for neuroinflammation. Thus, we suggest reserving the term neuroinflammation for contexts where multiple signs of inflammation are present to avoid erroneously classifying disorders as inflammatory when they may instead be caused by nonimmune etiologies or secondary immune processes that serve adaptive roles.
Collapse
|
3
|
Abstract
Inflammation occurs rapidly in response to acute brain insults such as stroke, haemorrhage or trauma, and can be sustained for long periods of time, for example in Alzheimer's or Parkinson's diseases and multiple sclerosis. Experimental evidence indicates that inflammation plays a major role in neurodegeneration under these conditions, and that the cytokine IL-1 (interleukin-1) is a pivotal mediator. IL-1 is expressed rapidly in response to neuronal injury, predominantly by microglia, and elevated levels of endogenous or exogenous IL-1 markedly exacerbate injury. The naturally occurring IL-1RA (IL-1 receptor antagonist) markedly inhibits ischaemic, excitotoxic and traumatic brain injury in rodents, and has shown promise in a Phase II clinical trial in stroke patients. The mechanisms of IL-1 expression, release and action in neurodegeneration are not fully elucidated and appear multiple. Systemic IL-1 markedly enhances ischaemic brain injury via release of neutrophils into circulation, neutrophil adhesion to injured cerebrovasculature and CNS (central nervous system) invasion, and cell death via activation of matrix metalloproteinase-9. IL-1 also influences the release of toxins from glial and endothelial cells. Neuronal responses to excitotoxins and physiological factors may have an impact on neuronal survival. IL-1RA, delivered peripherally, can enter the CNS in animals and humans and has no adverse effects in stroke or subarachnoid haemorrhage patients, but shows potential benefit in acute stroke patients.
Collapse
|
4
|
Patel HC, Ross FM, Heenan LE, Davies RE, Rothwell NJ, Allan SM. Neurodegenerative actions of interleukin-1 in the rat brain are mediated through increases in seizure activity. J Neurosci Res 2006; 83:385-91. [PMID: 16358339 DOI: 10.1002/jnr.20735] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The cytokine interleukin-1 (IL-1) is an established and important mediator of diverse forms of neuronal injury in experimental animals. However, its mechanisms of action remain largely unknown. We have reported previously that IL-1 markedly enhances excitotoxic injury induced in the rat by striatal administration of the excitotoxin alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA), leading to widespread neuronal loss throughout the ipsilateral cortex. Here we tested the hypothesis that IL-1 causes this injury through induction and/or enhancement of seizure activity in the rat. Consistently with this hypothesis, intrastriatal injection of AMPA or AMPA with IL-1 in the rat brain increased c-Fos expression in regions similar to those in which c-Fos has been reported previously in response to seizures. A significant increase in cortical neuronal activity (number of c-Fos positive cells) was observed in response to AMPA with IL-1 compared with AMPA (8 hr after injection). Increased seizure duration [3,522 +/- 660 sec (SEM) vs. 1,415 +/- 301 sec; P < 0.001] and cell death volume (140 +/- 20 mm3 vs. 52 +/- 6 mm3; P < 0.001) were seen in response to coinfusion of AMPA with IL-1 vs. AMPA alone. In addition, the anticonvulsant diazepam (intraperitoneal) significantly reduced cell death (P < 0.001) and seizure duration (P < 0.001) induced by AMPA with IL-1, and a significant correlation was found between seizure duration and cell death volume. These findings support our hypothesis that IL-1 enhances excitotoxic injury by enhancement of seizures, which may be of relevance to IL-1 actions in other forms of neuronal injury, including cerebral ischemia.
Collapse
Affiliation(s)
- H C Patel
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | | | | | | | | | | |
Collapse
|
5
|
Emsley HCA, Smith CJ, Georgiou RF, Vail A, Hopkins SJ, Rothwell NJ, Tyrrell PJ. A randomised phase II study of interleukin-1 receptor antagonist in acute stroke patients. J Neurol Neurosurg Psychiatry 2005; 76:1366-72. [PMID: 16170078 PMCID: PMC1739363 DOI: 10.1136/jnnp.2004.054882] [Citation(s) in RCA: 336] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The cytokine interleukin (IL)-1 mediates ischaemic brain damage in rodents. The endogenous, highly selective, IL-1 receptor antagonist (IL-1ra) protects against ischaemic cerebral injury in a range of experimental settings, and IL-1ra causes a marked reduction of cell death when administered peripherally or at a delay in transient cerebral ischaemia. We report here the first randomised, double blind, placebo controlled trial of recombinant human IL-1ra (rhIL-1ra) in patients with acute stroke. METHODS Patients within 6 hours of the onset of symptoms of acute stroke were randomised to rhIL-1ra or matching placebo. Test treatment was administered intravenously by a 100 mg loading dose over 60 seconds, followed by a 2 mg/kg/h infusion over 72 h. Adverse events and serious adverse events were recorded for up to 3 months, serial blood samples were collected for biological markers up to 3 months, and 5-7 day brain infarct volume was measured by computed tomography. RESULTS No adverse events were attributed to study treatment among 34 patients randomised. Markers of biological activity, including neutrophil and total white cell counts, C reactive protein, and IL-6 concentrations, were lower in rhIL-1ra treated patients. Among patients with cortical infarcts, clinical outcomes at 3 months in the rhIL-1ra treated group were better than in placebo treated. CONCLUSIONS These data suggest that rhIL-1ra is safe and well tolerated in acute stroke. In addition, rhIL-1ra exhibited biological activity that is relevant to the pathophysiology and clinical outcome of ischaemic stroke. Our findings identify rhIL-1ra as a potential new therapeutic agent for acute stroke.
Collapse
Affiliation(s)
- H C A Emsley
- Division of Neuroscience, The University of Liverpool, The Walton Centre for Neurology & Neurosurgery, Liverpool, UK
| | | | | | | | | | | | | |
Collapse
|
6
|
Hailer NP, Vogt C, Korf HW, Dehghani F. Interleukin-1β exacerbates and interleukin-1 receptor antagonist attenuates neuronal injury and microglial activation after excitotoxic damage in organotypic hippocampal slice cultures. Eur J Neurosci 2005; 21:2347-60. [PMID: 15932594 DOI: 10.1111/j.1460-9568.2005.04067.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The effects of interleukin (IL)-1beta and IL-1 receptor antagonist (IL-1ra) on neurons and microglial cells were investigated in organotypic hippocampal slice cultures (OHSCs). OHSCs obtained from rats were excitotoxically lesioned after 6 days in vitro by application of N-methyl-D-aspartate (NMDA) and treated with IL-1beta (6 ng/mL) or IL-1ra (40, 100 or 500 ng/mL) for up to 10 days. OHSCs were then analysed by bright field microscopy after hematoxylin staining and confocal laser scanning microscopy after labeling of damaged neurons with propidium iodide (PI) and fluorescent staining of microglial cells. The specificity of PI labeling of damaged neurons was validated by triple staining with neuronal and glial markers and it was observed that PI accumulated in damaged neurons only but not in microglial cells or astrocytes. Treatment of unlesioned OHSCs with IL-1beta did not induce neuronal damage but caused an increase in the number of microglial cells. NMDA lesioning alone resulted in a massive increase in the number of microglial cells and degenerating neurons. Treatment of NMDA-lesioned OHSCs with IL-1beta exacerbated neuronal cell death and further enhanced microglial cell numbers. Treatment of NMDA-lesioned cultures with IL-1ra significantly attenuated NMDA-induced neuronal damage and reduced the number of microglial cells, whereas application of IL-1ra in unlesioned OHSCs did not induce significant changes in either cell population. Our findings indicate that: (i) IL-1beta directly affects the central nervous system and acts independently of infiltrating hematogenous cells; (ii) IL-1beta induces microglial activation but is not neurotoxic per se; (iii) IL-1beta enhances excitotoxic neuronal damage and microglial activation and (iv) IL-1ra, even when applied for only 4 h, reduces neuronal cell death and the number of microglial cells after excitotoxic damage.
Collapse
Affiliation(s)
- Nils P Hailer
- Dr Senckenbergische Anatomie, Institute of Anatomy 2, Johann Wolfgang Goethe-University, D-60590 Frankfurt am Main, Federal Republic of Germany
| | | | | | | |
Collapse
|
7
|
Abstract
Injury to the central nervous system (CNS) and the resulting neuronal loss contribute significantly to morbidity and mortality in human and domestic animal populations. Most insults induce inflammation and the expression of cytokines. The specific roles of these proteins in neurological damage and repair are not completely understood. However, members of the interleukin-1 (IL-1) family have clear therapeutic potential: the IL-1 agonists, IL-1 alpha and IL-1 beta, are induced by CNS injury, and central injection of IL-1 increases, whilst peripheral or central administration of the IL-1 antagonist, IL-1ra, reduces the extent of the damage by more than 50%. The mechanism of action of these cytokines is the subject of intense research. In this review, we summarise approaches that are being used to investigate neuronal cell death, and the contribution of inflammation and cytokines, in particular the IL-1 family, to neurodegenerative disorders.
Collapse
Affiliation(s)
- Rosemary M Gibson
- School of Biological Sciences, 1.124 Stopford Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | | | | |
Collapse
|
8
|
Cowen MS, Schroff KC, Gass P, Sprengel R, Spanagel R. Neurobehavioral effects of alcohol in AMPA receptor subunit (GluR1) deficient mice. Neuropharmacology 2003; 45:325-33. [PMID: 12871650 DOI: 10.1016/s0028-3908(03)00174-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Of the ionotropic glutamatergic receptors, the NMDA receptor is clearly implicated in the acute and chronic effects of ethanol; however, the role of the AMPA receptor in mediating the effects of ethanol in vivo is as yet unclear. Using mice deficient in the AMPA receptor subunit GluR1 (GluR1-/- mice), we investigated whether the AMPA receptor had a significant role in mediating the effects of ethanol. GluR1-/- mice showed greater locomotor activity in a novel environment, but by the fifth day of repeated testing their activity was the same as that of wild-type mice. In contrast to their enhanced locomotor activity, on an accelerating rotarod GluR1-/- mice performed consistently worse than wild-types. With regard to the effects of ethanol on motor responses, GluR1-/- mice did not differ significantly from wild-type mice in ethanol's sedative or incoordinating effects. However, the GluR1-/- mice were insensitive to the hypothermic effects of a hypnotic dose of ethanol in contrast to wild-types; this effect was dissociable from the hypnotic effects of ethanol. Further, tolerance to ethanol developed equally for GluR1-/- mice versus wild-type mice. In terms of alcohol drinking behavior, compared to wild-types, GluR1-/- mice differed neither in the acquisition of voluntary ethanol consumption nor in stress-induced ethanol drinking, nor in the expression of an alcohol deprivation effect (ADE) which is used as a model of relapse-like drinking behavior. In summary, although the loss of a hypothermic effect of ethanol in GluR1-/- mice indicates a critical role for the AMPA receptors in this effect, the GluR1 subunit of the AMPA receptor does not seem to play a critical role in the etiology of alcohol dependence. However, changes observed in activity patterns may be related to the putative role of AMPA receptors in attention deficit hyperactivity disorder.
Collapse
Affiliation(s)
- M S Cowen
- Department of Psychopharmacology, University of Heidelberg, Central Institute of Mental Health (CIMH), J5, 68159, Mannheim, Germany
| | | | | | | | | |
Collapse
|
9
|
Halladay AK, Kusnecov A, Michna L, Kita T, Hara C, Wagner GC. Relationship between methamphetamine-induced dopamine release, hyperthermia, self-injurious behaviour and long term dopamine depletion in BALB/c and C57BL/6 mice. PHARMACOLOGY & TOXICOLOGY 2003; 93:33-41. [PMID: 12828572 DOI: 10.1034/j.1600-0773.2003.930105.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Differential sensitivity to neurotoxic effects of methamphetamine on striatal dopaminergic neurones between C57BL/6 and BALB/c mice has been established. In the present studies, the interaction of methamphetamine-induced dopamine release, self-injurious behaviour, the neural immune response, and the long-term (3 day) dopamine depletion were examined in these strains after administration of 8 mg/kg methamphetamine. BALB/c mice showed increased hyperthermia compared to the C57BL/6 strain, as well as induction of interleukin-1beta. Additionally, homovanillic acid (HVA) levels, as well as HVA/DA turnover ratios were elevated in the striatum and frontal cortex of BALB/c mice, both compared to untreated mice and to the C57BL/6 strain after a single injection of methamphetamine. Pretreatment with acetaminophen eliminated the methamphetamine-induced hyperthermia in BALB/c mice and reduced body temperature in C57BL/6 mice. However, acetaminophen pretreatment did not affect any parameters of dopaminergic toxicity in the striatum or frontal cortex of the BALB/c strain following repeated methamphetamine injections. Furthermore, acetaminophen pretreatment did not alter the incidence of self-injurious behaviour in BALB/c mice. Therefore, hyperthermia and methamphetamine-induced toxicity appear to be independent phenomena while self-injurious behaviour may provide a better predictor of toxicity, which, in turn, may be related to dopamine release.
Collapse
Affiliation(s)
- Alycia K Halladay
- Joint Graduate Program in Toxicology, Rutgers University, Piscataway, NJ 08854, USA
| | | | | | | | | | | |
Collapse
|
10
|
Ma XC, Gottschall PE, Chen LT, Wiranowska M, Phelps CP. Role and mechanisms of interleukin-1 in the modulation of neurotoxicity. Neuroimmunomodulation 2003; 10:199-207. [PMID: 12584407 DOI: 10.1159/000068322] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2001] [Accepted: 05/23/2002] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Recent studies on cerebral ischemia in the rat have demonstrated that administration of interleukin-1 receptor antagonist (IL-1ra) markedly reduces the volumes of infarcts which are associated with N-methyl-D-aspartate (NMDA)-mediated neurotoxicity. These observations suggested that endogenous interleukin-1 (IL-1) may be involved in the mediation of excitotoxic neuronal injury following ischemia. METHOD In the present studies, we examined the role of interleukin-1beta (IL-1beta) in NMDA-related and microglia-induced excitotoxicity in cocultures of mixed neurons and microglia. RESULTS Our observations in these mixed cultures indicated that addition of IL-1beta exaggerated NMDA and glutamate-evoked hippocampal neuron death. Addition of microglia, activated by lipopolysaccharide (LPS) and interferon-gamma (IFN-gamma), to cocultures of cortical neurons and glia induced significantly greater neurotoxicity when compared with cocultures of cortical neurons and untreated microglia. This neurotoxicity did not require that activated glia be in cell-to-cell contact with neurons. Addition of either IL-1ra or the NMDA receptor antagonist MK-801 to cocultures of cortical neurons and activated glia partially reversed the neuronal damage mediated by activated microglia. Finally, IL-1beta concentrations in the supernatant of cocultures of cortical neurons and microglia treated by LPS and IFN-gamma were markedly increased when compared with coculture of neurons with untreated microglia. CONCLUSION These results suggest that both the NMDA receptor and the IL-1 receptor are involved in microglia-mediated neurotoxicity.
Collapse
Affiliation(s)
- Xiaoling C Ma
- Department of Anatomy, University of South Florida, Tampa, Florida 336124-4799, USA
| | | | | | | | | |
Collapse
|
11
|
Haspolat S, Mihçi E, Coşkun M, Gümüslü S, Ozben T, Yeğin O, Ozbenm T. Interleukin-1beta, tumor necrosis factor-alpha, and nitrite levels in febrile seizures. J Child Neurol 2002; 17:749-51. [PMID: 12546429 DOI: 10.1177/08830738020170101501] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Proinflammatory cytokines (such as interleukin-1beta, tumor necrosis factor-alpha) and nitric oxide are known to have both direct and indirect modulating effects on neurons and neurotoxic neurotransmitters released during excitation or inflammation. We measured interleukin-1beta, tumor necrosis factor-alpha, and nitrite levels in the peripheral blood and cerebrospinal fluid of children with febrile seizures and compared our results with those of children with febrile illnesses without seizures. Twenty-nine children with febrile seizure and 15 controls were studied. The mean concentrations of interleukin-1beta and nitrite were significantly increased in the cerebrospinal fluid (P < .01) of the children with febrile seizure. There were no significant changes in serum interleukin-1beta, tumor necrosis factor-alpha, nitrite, and cerebrospinal fluid tumor necrosis factor-alpha levels. Our data support the hypothesis that increased production of interleukin-1beta in the central nervous system or increased diffusion of interleukin-1beta through the blood-brain barrier is involved in the pathogenesis of febrile seizures.
Collapse
Affiliation(s)
- Senay Haspolat
- Department of Pediatric Neurology, Akdeniz University Medical School, Antalya, Turkey.
| | | | | | | | | | | | | |
Collapse
|
12
|
Grundy RI, Rothwell NJ, Allan SM. Site-specific actions of interleukin-1 on excitotoxic cell death in the rat striatum. Brain Res 2002; 926:142-8. [PMID: 11814416 DOI: 10.1016/s0006-8993(01)03325-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The pro-inflammatory cytokine interleukin-1 (IL-1) contributes to and exacerbates many forms of neurodegeneration. When co-administered with the potent glutamatergic agonist S-alpha-amino-3-hydroxy-5-methyl-4-isoxazole-propionate (S-AMPA) in the rat striatum, IL-1 induces marked and widespread cell death throughout the ipsilateral cortex. The mechanisms underlying this action of IL-1 are not known but may involve activation of polysynaptic neuronal pathways leading from the striatum to the cortex via other brain areas such as the hypothalamus. The aims of the present study were to identify specific sites of action of IL-1 in the rat striatum, in order to further understand these pathways. Ventral regions of the caudate-putamen and the lateral shell of the nucleus accumbens (NAcc) were particularly sensitive to the effects of IL-1 on excitotoxic damage. A high percentage of co-injections in these sites induced distant cortical damage, whereas injections in more dorsal areas of the caudate-putamen or core regions of the NAcc were less likely to result in cortical cell death. The 'positive' injection sites differ from the unresponsive areas in that they have extensive connections with the limbic system and it may be that IL-1 displays specific actions on limbic pathways that, in conjunction with AMPA/kainate receptor activation, contribute to the remote cell death in the cortex. These findings enhance our understanding of the actions of IL-1, and the mechanisms by which it participates in neurodegeneration through both local and long-range effects.
Collapse
Affiliation(s)
- Robert I Grundy
- Schering-Plough Research Institute, San Raffaele Science Park, Via Olgettina 58, 20132 Milan, Italy
| | | | | |
Collapse
|
13
|
Abstract
Experimental and clinical damage to the brain leads to rapid upregulation of an array of cytokines predominantly by glia. These cytokines may exert neurotoxic or neuroprotective actions. This paper will focus on the pro-inflammatory cytokine interleukin-1 (IL-1), which participates in diverse forms of brain damage including ischemia, brain trauma, and excitotoxic injury. Administration of low doses of IL-1 markedly exacerbates these forms of brain damage, whereas blocking IL-1 release or actions reduces neuronal death. IL-1 receptor antagonist (IL-1ra) is also upregulated by brain damage (mainly by neurons) and acts as an endogenous inhibitor of neurodegeneration, presumably by blocking IL-1 actions on its receptor. We have studied the actions of both IL-1 and IL-1ra in experimental models of ischemic and neurotoxic injury in rats, and have found site-specific effects within the striatum. On the basis of this and further work, we propose that IL-1 can exacerbate cell death in these conditions by modifying polysynaptic anterograde pathways leading from the striatum to the cortex. The precise nature of these pathways remains undetermined, as do the underlying mechanisms by which IL-1 can exert its effects, but appear to involve induction of IL-1 in specific brain regions and activation of cortical glutamatergic pathways.
Collapse
Affiliation(s)
- S M Allan
- School of Biological Sciences, 1.124 Stopford Building, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom.
| |
Collapse
|
14
|
Dommergues MA, Patkai J, Renauld JC, Evrard P, Gressens P. Proinflammatory cytokines and interleukin-9 exacerbate excitotoxic lesions of the newborn murine neopallium. Ann Neurol 2001. [DOI: 10.1002/1531-8249(200001)47:1<54::aid-ana10>3.0.co;2-y] [Citation(s) in RCA: 162] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
15
|
Allan SM, Parker LC, Collins B, Davies R, Luheshi GN, Rothwell NJ. Cortical cell death induced by IL-1 is mediated via actions in the hypothalamus of the rat. Proc Natl Acad Sci U S A 2000; 97:5580-5. [PMID: 10779559 PMCID: PMC25871 DOI: 10.1073/pnas.090464197] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The cytokine IL-1 mediates diverse forms of neurodegeneration, but its mechanism of action is unknown. We have demonstrated previously that exogenous and endogenous IL-1 acts specifically in the rat striatum to dramatically enhance ischemic and excitotoxic brain damage and cause extensive cortical injury. Here we tested the hypothesis that this distant effect of IL-1 is mediated through polysynaptic striatal outputs to the cortex via the hypothalamus. We show that IL-1beta injected into the rat striatum with the excitotoxin alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (S-AMPA) caused increased expression of IL-1beta (mRNA and protein) mainly in the cortex where maximum injury occurs. Marked increases in IL-1beta mRNA and protein were also observed in the hypothalamus. S-AMPA, injected alone into the striatum, caused only localized damage, but administration of IL-1beta into either the striatum or the lateral hypothalamus immediately after striatal S-AMPA resulted in widespread cell loss throughout the ipsilateral cortex. Finally we showed that the cortical cell death produced by striatal coinjection of S-AMPA and IL-1beta was significantly reduced by administration of the IL-1 receptor antagonist into the lateral hypothalamus. These data suggest that IL-1beta can act in the hypothalamus to modify cell viability in the cortex. We conclude that IL-1-dependent pathways project from the striatum to the cortex via the hypothalamus and lead to cortical injury, and that these may contribute to a number of human neurological conditions including stroke and head trauma.
Collapse
Affiliation(s)
- S M Allan
- School of Biological Sciences, 1.124 Stopford Building, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom
| | | | | | | | | | | |
Collapse
|