51
|
Radiolabeling with fluorine-18 of a protein, interleukin-1 receptor antagonist. Appl Radiat Isot 2010; 68:1721-7. [PMID: 20435481 DOI: 10.1016/j.apradiso.2010.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Revised: 02/26/2010] [Accepted: 04/01/2010] [Indexed: 11/23/2022]
Abstract
IL-1RA is a naturally occurring antagonist of the pro-inflammatory cytokine interleukin-1 (IL-1) with high therapeutic promise, but its pharmacokinetic remains poorly documented. In this report, we describe the radiolabeling of recombinant human interleukin-1 receptor antagonist (rhIL-1RA) with fluorine-18 to allow pharmacokinetic studies by positron emission tomography (PET). rhIL-1RA was labeled randomly by reductive alkylation of free amino groups (the epsilon-amino group of lysine residues or amino-terminal residues) using [(18)F]fluoroacetaldehyde under mild reaction conditions. Radiosyntheses used a remotely controlled experimental rig within 100min and the radiochemical yield was in the range 7.1-24.2% (decay corrected, based on seventeen syntheses). We showed that the produced [(18)F]fluoroethyl-rhIL-1ra retained binding specificity by conducting an assay on rat brain sections, allowing its pharmakokinetic study using PET.
Collapse
|
52
|
Diestel A, Troeller S, Billecke N, Sauer IM, Berger F, Schmitt KRL. Mechanisms of hypothermia-induced cell protection mediated by microglial cellsin vitro. Eur J Neurosci 2010; 31:779-87. [DOI: 10.1111/j.1460-9568.2010.07128.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
53
|
Interleukin-1 receptor antagonist inhibits the release of glutamate, hydroxyl radicals, and prostaglandin E2 in the hypothalamus during pyrogen-induced fever in rabbits. Eur J Pharmacol 2010; 629:125-31. [DOI: 10.1016/j.ejphar.2009.11.060] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Revised: 11/11/2009] [Accepted: 11/23/2009] [Indexed: 11/22/2022]
|
54
|
Pluta R, Ułamek M, Jabłoński M. Alzheimer's mechanisms in ischemic brain degeneration. Anat Rec (Hoboken) 2010; 292:1863-81. [PMID: 19943340 DOI: 10.1002/ar.21018] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
There is increasing evidence for influence of Alzheimer's proteins and neuropathology on ischemic brain injury. This review investigates the relationships between beta-amyloid peptide, apolipoproteins, presenilins, tau protein, alpha-synuclein, inflammation factors, and neuronal survival/death decisions in brain following ischemic episode. The interactions of these molecules and influence on beta-amyloid peptide synthesis and contribution to ischemic brain degeneration and finally to dementia are reviewed. Generation and deposition of beta-amyloid peptide and tau protein pathology are important key players involved in mechanisms in ischemic neurodegeneration as well as in Alzheimer's disease. Current evidence suggests that inflammatory process represents next component, which significantly contribute to degeneration progression. Although inflammation was initially thought to arise secondary to ischemic neurodegeneration, recent studies present that inflammatory mediators may stimulate amyloid precursor protein metabolism by upregulation of beta-secretase and therefore are able to establish a vicious cycle. Functional brain recovery after ischemic lesion was delayed and incomplete by an injury-related increase in the amount of the neurotoxic C-terminal of amyloid precursor protein and beta-amyloid peptide. Moreover, ischemic neurodegeneration is strongly accelerated with aging, too. New therapeutic alternatives targeting these proteins and repairing related neuronal changes are under development for the treatment of ischemic brain consequences including memory loss prevention.
Collapse
Affiliation(s)
- Ryszard Pluta
- Laboratory of Ischemic and Neurodegenerative Brain Research, Department of Neurodegenerative Disorders, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5 Str., Warsaw, Poland.
| | | | | |
Collapse
|
55
|
Luheshi NM, Rothwell NJ, Brough D. Dual functionality of interleukin-1 family cytokines: implications for anti-interleukin-1 therapy. Br J Pharmacol 2010; 157:1318-29. [PMID: 19681864 PMCID: PMC2765320 DOI: 10.1111/j.1476-5381.2009.00331.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Dysregulated inflammation contributes to disease pathogenesis in both the periphery and the brain. Cytokines are coordinators of inflammation and were originally defined as secreted mediators, released from expressing cells to activate plasma membrane receptors on responsive cells. However, a group of cytokines is now recognized as having dual functionality. In addition to their extracellular effects, these cytokines act inside the nuclei of cytokine-expressing or cytokine-responsive cells. Interleukin-1 (IL-1) family cytokines are key pro-inflammatory mediators, and blockade of the IL-1 system in inflammatory diseases is an attractive therapeutic goal. All current therapies target IL-1 extracellular actions. Here we review evidence that suggests IL-1 family members have dual functionality. Several IL-1 family members have been detected inside the nuclei of IL-1-expressing or IL-1-responsive cells, and intranuclear IL-1 is reported to regulate gene transcription and mRNA splicing. However, further work is required to determine the impact of IL-1 intranuclear actions on disease pathogenesis. The intranuclear actions of IL-1 family members represent a new and potentially important area of IL-1 biology and may have implications for the future development of anti-IL-1 therapies.
Collapse
Affiliation(s)
- N M Luheshi
- Faculty of Life Sciences, University of Manchester, Manchester, UK.
| | | | | |
Collapse
|
56
|
Plasma interleukin-1beta concentration is associated with stroke in sickle cell disease. Cytokine 2009; 49:39-44. [PMID: 19900820 DOI: 10.1016/j.cyto.2009.10.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2008] [Revised: 05/11/2009] [Accepted: 10/06/2009] [Indexed: 12/15/2022]
Abstract
The pathogenesis of sickle cell disease (HbSS), which has numerous complications including stroke, involves inflammation resulting in alteration of plasma inflammatory protein concentration. We investigated HbSS children with abnormal cerebral blood flow detected by trans-cranial Doppler ultrasound (TCD) who participated in the multi-center stroke prevention (STOP) study, to determine if plasma inflammatory protein concentration is associated with the outcome of stroke. Thirty-nine plasma samples from HbSS participants with elevated TCD who had no stroke, HbSS-NS (n=13) or had stroke, HbSS-S (n=13), HbSS steady-state controls (n=7) and controls with normal hemoglobin, HbAA (n=6), were analyzed simultaneously for 27 circulating inflammatory proteins. Logistic regression and receiver operating characteristics curve analysis of stroke on plasma inflammatory mediator concentration, adjusted for age and gender, demonstrated that interleukin-1beta (IL-1beta) was protective against stroke development (HbSS-NS=19, 17-23, HbSS-S=17, 16-19 pg/mL, median and 25th-75th percentile; odds ratio=0.59, C.I.=0.36-0.96) and was a good predictor of stroke (area under curve=0.852). This result demonstrates a strong association of systemic inflammation with stroke development in HbSS via moderately increased plasma IL-1beta concentration, which is furthermore associated with a decreased likelihood of stroke in HbSS.
Collapse
|
57
|
McNamee EN, Ryan KM, Kilroy D, Connor TJ. Noradrenaline induces IL-1ra and IL-1 type II receptor expression in primary glial cells and protects against IL-1beta-induced neurotoxicity. Eur J Pharmacol 2009; 626:219-28. [PMID: 19818755 DOI: 10.1016/j.ejphar.2009.09.054] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Revised: 09/18/2009] [Accepted: 09/28/2009] [Indexed: 10/20/2022]
Abstract
The pro-inflammatory cytokine interleukin-1beta (IL-1beta) plays a key role in initiating an immune response within the central nervous system (CNS), and is thought to be a significant contributor to the neurodegenerative process. The actions of IL-1beta can be regulated by interleukin-1 receptor antagonist (IL-1ra), which prevents IL-1beta from acting on the IL-1 type I receptor (IL-1RI). Another negative regulator of the IL-1 system is the IL-1 type II receptor (IL-1RII); a decoy receptor that serves to sequester IL-1. Consequently, pharmacological strategies that tip the balance in favour of IL-1ra and IL-1RII may be of therapeutic benefit. Evidence suggests that the neurotransmitter noradrenaline elicits anti-inflammatory actions in the CNS, and consequently may play an endogenous neuroprotective role. Here we report that noradrenaline induces production of IL-1ra and IL-1RII from primary rat mixed glial cells. In contrast, noradrenaline did not alter IL-1beta expression, or expression of IL-1RI or the IL-1 type I receptor accessory protein (IL-1RAcp); both of which are required for IL-1 signalling. Our results demonstrate that the ability of noradrenaline to induce IL-1ra and IL-1RII is mediated via beta-adrenoceptor activation and downstream activation of protein kinase A and extracellular signal-regulated kinase (ERK). In parallel with its ability to increase IL-1ra and IL-1RII, noradrenaline prevented neurotoxicity in cortical primary neurons induced by conditioned medium from IL-1beta treated mixed glial cells. These data indicate that noradrenaline negatively regulates IL-1 system in glial cells and has neuroprotective properties in situations where IL-1 contributes to pathology.
Collapse
Affiliation(s)
- Eoin N McNamee
- Neuroimmunology Research Group, Department of Physiology, School of Medicine & Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | | | | | | |
Collapse
|
58
|
Pan XD, Chen XC, Zhu YG, Chen LM, Zhang J, Huang TW, Ye QY, Huang HP. Tripchlorolide protects neuronal cells from microglia-mediated β-amyloid neurotoxicity through inhibiting NF-κB and JNK signaling. Glia 2009; 57:1227-38. [DOI: 10.1002/glia.20844] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
59
|
Ischemic preconditioning-induced neuroprotection is associated with differential expression of IL-1beta and IL-1 receptor antagonist in the ischemic cortex. J Neuroimmunol 2009; 217:14-9. [PMID: 19545912 DOI: 10.1016/j.jneuroim.2009.06.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Revised: 05/22/2009] [Accepted: 06/01/2009] [Indexed: 02/03/2023]
Abstract
Ischemic preconditioning (IP) is a phenomenon that organs develop a tolerance toward subsequent lethal ischemic insults. Among the factors that are involved in IP, IL-1beta and its endogenous receptor antagonist IL-1ra have been identified as important players in the induction of IP. The present study investigated whether IP affects the levels of these two antagonistic proteins during tolerance and reperfusion periods after ischemic stroke. The IP 24 h prior to ischemic stroke resulted in neuroprotection in the cortex. IP-induced protection is accompanied by increased IL-1beta gene and IL-1ra gene and protein levels during the tolerance period. In the post-ischemic cortex, IP resulted in the suppression of IL-1beta mRNA and protein levels at 6 h without affecting IL-1ra expression and the up-regulation of IL-1ra protein at 24 h. These findings demonstrate that IP differentially regulates cortical IL-1beta and IL-1ra expression before and after ischemic stroke and suggest that the shift toward an anti-inflammatory state in the post-ischemic cortex may contribute to IP-induced neuroprotection.
Collapse
|
60
|
Spulber S, Bartfai T, Schultzberg M. IL-1/IL-1ra balance in the brain revisited - evidence from transgenic mouse models. Brain Behav Immun 2009; 23:573-9. [PMID: 19258032 DOI: 10.1016/j.bbi.2009.02.015] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2008] [Revised: 02/09/2009] [Accepted: 02/19/2009] [Indexed: 01/07/2023] Open
Abstract
The interleukin-1 (IL-1) family is unique in its including an endogenous antagonist of the IL-1 receptor (IL-1ra). IL-1ra has been shown to antagonise IL-1 signalling so effectively, that it came into clinical use within a few years from its discovery. Although barely detectable in the normal brain, IL-1 is dramatically upregulated during neuroinflammation, and also displays peaks of expression in the brain during development, as well as following the induction of long-term potentiation. IL-1 has been ascribed a central role in neuroinflammation accompanying ageing and age-related neurodegenerative conditions. Several experimental models based on genetically modified mice have been used in order to address the role of IL-1 in neurodegeneration and neuroprotection. Most of the findings here are based on the experiments involving a transgenic mouse strain with brain-directed overexpression of human IL-1ra, in which the balance between IL-1 and IL-1ra is permanently tipped towards inhibiting IL-1 signalling. The developmental effects of IL-1 are evident in the altered brain morphology in adult transgenic mice. In addition, IL-1 appears to be central in regulating the elasticity of the brain response to injury. Thus, a number of lines of evidence support the essential role played by IL-1 in development, plasticity, and physiological brain function.
Collapse
Affiliation(s)
- S Spulber
- Karolinska Institutet, Dept. of Neurobiology, Care Sciences and Society, Stockholm, Sweden.
| | | | | |
Collapse
|
61
|
Girard S, Kadhim H, Roy M, Lavoie K, Brochu ME, Larouche A, Sébire G. Role of perinatal inflammation in cerebral palsy. Pediatr Neurol 2009; 40:168-74. [PMID: 19218029 DOI: 10.1016/j.pediatrneurol.2008.09.016] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 08/07/2008] [Accepted: 09/17/2008] [Indexed: 01/19/2023]
Abstract
Inflammatory molecules are promptly upregulated in the fetal environment and postnatally in brain-damaged subjects. Intrauterine infections and inflammation are often associated with asphyxia. This double-hit effect by combined infection or inflammation and hypoxia is therefore a frequent concomitant in neonatal brain damage. Animal models combining hypoxia and infection were recently designed to explore the mechanisms underlying brain damage in such circumstances and to look for possible neuroprotective strategies. Proinflammatory cytokines are thought to be major mediators in brain injury in neonates with perinatal asphyxia, bacterial infection, or both. Cytokines, however, could also have neuroprotective properties. The critical point in the balance between neurodamaging and neuroprotective effects of cytokines has yet to be unraveled. This understanding might help to develop new therapeutic approaches to counteract the inflammatory disequilibrium observed in the pathophysiologic mechanisms associated with brain injury.
Collapse
Affiliation(s)
- Sylvie Girard
- Laboratory of Neuropediatrics-Laboratoire de Neuropédiatrie, Neurosciences Centre, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
62
|
Vogt C, Hailer NP, Ghadban C, Korf HW, Dehghani F. Successful inhibition of excitotoxic neuronal damage and microglial activation after delayed application of interleukin-1 receptor antagonist. J Neurosci Res 2009; 86:3314-21. [PMID: 18646209 DOI: 10.1002/jnr.21792] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Interleukin (IL)-1 is an important mediator of neuronal demise and glial activation after acute central nervous system lesions and is antagonized by IL-1 receptor antagonist (IL-1RA). Here we determined the time window in which IL-1RA elicits neuroprotective effects in rat organotypic hippocampal slice cultures (OHSC). OHSC were lesioned with N-methyl-D-aspartate (NMDA) and treated with IL-1RA (100 ng/ml) at different time points postinjury or were left untreated. Damaged neurons, microglial cells, and astrocytes were labelled with NeuN, propidium iodide, isolectin B(4), or glial fibrillary acidic protein (GFAP), respectively, and were analyzed by confocal laser scanning microscopy. In lesioned OHSC, the most dramatic increase in microglial cell number occurred between 8 and 16 hr postinjury, and the maximal neuronal demise was found between 16 and 24 hr postinjury. The cellular source of IL-1beta was investigated by immunohistochemistry, and IL-1beta immunoreactivity was found in few microglial cells at 4 hr postinjury and in numerous microglial cells and astrocytes at 16 hr postinjury. In both glial populations, IL-1beta immunoreactivity peaked at 24 hr postinjury. IL-1RA treatment potently suppressed neuronal damage by 55% when initiated within the first 16 hr postinjury (P < 0.05), and IL-1RA treatment initiated at 24 hr postinjury resulted in weaker but still significant neuroprotection. IL-1RA treatment also reduced the number of microglial cells significantly when initiated within 36 hr postinjury (P < 0.05). In conclusion, IL-1RA exhibits significant neuroprotective effects in this in vitro model of excitotoxic injury even after delayed application.
Collapse
Affiliation(s)
- Cornelia Vogt
- Dr. Senckenbergische Anatomie, Institut für Anatomie II, Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany
| | | | | | | | | |
Collapse
|
63
|
Pinteaux E, Trotter P, Simi A. Cell-specific and concentration-dependent actions of interleukin-1 in acute brain inflammation. Cytokine 2008; 45:1-7. [PMID: 19026559 DOI: 10.1016/j.cyto.2008.10.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2008] [Revised: 09/15/2008] [Accepted: 10/10/2008] [Indexed: 01/03/2023]
Abstract
Interleukin (IL)-1 is a pivotal pro-inflammatory cytokine and an important mediator of both acute and chronic central nervous system (CNS) injuries. Despite intense research in CNS IL-1 biology over the past two decades, its precise mechanism of action in inflammatory responses to acute brain disorders remains largely unknown. In particular, much effort has been focussed on using in vitro approaches to better understand the cellular and signalling mechanisms of actions of IL-1, yet some discrepancies in the literature regarding the effects produced by IL-1beta in in vitro paradigms of injury still exist, particularly as to whether IL-1 exerts neurotoxic or neuroprotective effects. Here we aim to review the cell-specific and concentration-dependent actions of IL-1 in brain cells, to depict the mechanism by which this cytokine induces neurotoxicity or neuroprotection in acute brain injury.
Collapse
Affiliation(s)
- Emmanuel Pinteaux
- Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom.
| | | | | |
Collapse
|
64
|
Degos V, Teissier N, Gressens P, Puybasset L, Mantz J. [Inflammation and acute brain injuries in intensive care]. ACTA ACUST UNITED AC 2008; 27:1008-15. [PMID: 19010639 DOI: 10.1016/j.annfar.2008.07.099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Accepted: 07/17/2008] [Indexed: 11/26/2022]
Abstract
Patients with acute brain injuries or susceptibility to post-surgery stroke are a major therapeutic challenge for intensive care and anaesthesiology medicine. The control of systemic stress involved in brain damage is necessary to reduce the frequency and severity of secondary brain lesions. Inflammation is known to be directly involved in acute brain lesions. The brain is a major participant in inflammation control through activation or inhibition effects. The exact mechanisms involved in deleterious effects following acute brain injuries due to inflammation are still unknown. This non-exhaustive study will expose the principal processes involved in inflammatory brain disease and explain the consequences of peripheral inflammation for the brain. Neuroprotection strategies in acute neuroinflammation will be reported with a focus on anaesthetic agents and the inflammation cascade.
Collapse
Affiliation(s)
- V Degos
- Unité Inserm U676, hôpital Robert-Debré, 48, boulevard Serrurier, 75019 Paris, France.
| | | | | | | | | |
Collapse
|
65
|
Fiedorowicz A, Figiel I, Zaremba M, Dzwonek K, Schliebs R, Oderfeld-Nowak B. Trimethyltin-evoked apoptosis of murine hippocampal granule neurons is accompanied by the expression of interleukin-1beta and interleukin-1 receptor antagonist in cells of ameboid phenotype, the majority of which are NG2-positive. Brain Res Bull 2008; 77:19-26. [DOI: 10.1016/j.brainresbull.2008.02.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Accepted: 02/06/2008] [Indexed: 01/02/2023]
|
66
|
Wanderer AA. Ischemic-reperfusion syndromes: Biochemical and immunologic rationale for IL-1 targeted therapy. Clin Immunol 2008; 128:127-32. [DOI: 10.1016/j.clim.2008.03.514] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2008] [Revised: 03/19/2008] [Accepted: 03/22/2008] [Indexed: 11/28/2022]
|
67
|
Pan XD, Chen XC, Zhu YG, Zhang J, Huang TW, Chen LM, Ye QY, Huang HP. Neuroprotective role of tripchlorolide on inflammatory neurotoxicity induced by lipopolysaccharide-activated microglia. Biochem Pharmacol 2008; 76:362-72. [DOI: 10.1016/j.bcp.2008.05.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2008] [Revised: 05/10/2008] [Accepted: 05/13/2008] [Indexed: 11/26/2022]
|
68
|
Rusai K, Huang H, Sayed N, Strobl M, Roos M, Schmaderer C, Heemann U, Lutz J. Administration of interleukin-1 receptor antagonist ameliorates renal ischemia-reperfusion injury. Transpl Int 2008; 21:572-80. [DOI: 10.1111/j.1432-2277.2008.00651.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
69
|
Chen G, Frøkiaer J, Pedersen M, Nielsen S, Si Z, Pang Q, Stødkilde-Jørgensen H. Reduction of ischemic stroke in rat brain by alpha melanocyte stimulating hormone. Neuropeptides 2008; 42:331-8. [PMID: 18359516 DOI: 10.1016/j.npep.2008.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Revised: 01/22/2008] [Accepted: 01/25/2008] [Indexed: 11/16/2022]
Abstract
Anti-inflammatory therapy has provided hope for a new effective treatment of brain ischemic stroke. In this study, adult male Wistar rats subjected to right middle cerebral artery occlusion (MCAO) for 60 min were allocated to treatment of the anti-inflammatory compound alpha melanocyte stimulating hormone (alpha-MSH) or saline. Magnetic resonance imaging (MRI) and histology were used to evaluate the effects of alpha-MSH. MRI volumetry was performed to measure infarct size, and MRI measurements of the apparent diffusion coefficient (ADC) were performed to evaluate changes in the extra/intracellular volume ratio. Triphenyltetrazolium chloride (TTC) staining was used as a reference method to measure infarct sizes. The ADC value of the infarct area decreased significantly two days after MCAO in both groups. Simultaneously the infarct volume determined from the ADC map decreased in the alpha-MSH treated group compared to the control group. Five days after MCAO, ADC returned to baseline levels in both groups. The infarct volume in the alpha-MSH group was smaller compared to the saline treated group as demonstrated both by MRI and TTC staining. This study showed that the extra/intracellular ratio (reflected by ADC) following focal brain ischemic stroke could be affected by alpha-MSH. Secondly, we showed that the infarct volume was reduced by alpha-MSH. The volumetric dimensions of the infarct areas measured by MRI were comparable to those measured by histology.
Collapse
Affiliation(s)
- Gang Chen
- MR Research Centre, University of Aarhus, Aarhus University Hospital, Skejby, Aarhus N, Denmark.
| | | | | | | | | | | | | |
Collapse
|
70
|
Abstract
Multiple lines of evidence suggest that inflammation and glutamate dysfunction contribute to the pathophysiology of depression. In this review we provide an overview of how these two systems may interact. Excess levels of inflammatory mediators occur in a subgroup of depressed patients. Studies of acute experimental activation of the immune system with endotoxin and of chronic activation during interferon-alpha treatment show that inflammation can cause depression. Peripheral inflammation leads to microglial activation which could interfere with excitatory amino acid metabolism leading to inappropriate glutamate receptor activation. Loss of astroglia, a feature of depression, upsets the balance of anti- and pro-inflammatory mediators and further impairs the removal of excitatory amino acids. Microglia activated by excess inflammation, astroglial loss, and inappropriate glutamate receptor activation ultimately disrupt the delicate balance of neuroprotective versus neurotoxic effects in the brain, potentially leading to depression.
Collapse
|
71
|
Shen KH, Chang CK, Lin MT, Chang CP. Interleukin-1 receptor antagonist restores homeostatic function and limits multiorgan damage in heatstroke. Eur J Appl Physiol 2008; 103:561-8. [DOI: 10.1007/s00421-008-0755-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2008] [Indexed: 10/22/2022]
|
72
|
Acute neurodegeneration and the inflammasome: central processor for danger signals and the inflammatory response? J Cereb Blood Flow Metab 2008; 28:867-81. [PMID: 18212795 DOI: 10.1038/sj.jcbfm.9600609] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Activation of the inflammatory response is a crucial event in the adverse outcome of cerebral ischemia, which is promoted by proinflammatory cytokines such as interleukin (IL)-1beta. Although caspase-1 is necessary for IL-1beta processing, the 'upstream' signaling pathways were, until recently, essentially unknown. Fortunately, the inflammasome, a multiprotein complex responsible for activating caspase-1 and caspase-5, has recently been characterized. The activation of the inflammasome can result in one of several consequences such as cytokine secretion, cell death, or the development of a stress-resistant state. The significance of the inflammasome for the initiation of the inflammatory response during systemic diseases has already been shown and members of the inflammasome complex were recently found to be induced in acute brain injury. However, the specific pathophysiologic role of the inflammasome in neurodegenerative disorders still remains to be clarified. The underlying theories (e.g., danger signal theory) along with the signaling pathways that link the inflammasome to acute neurodegeneration will be discussed here. Furthermore, the stimuli that potentially activate the inflammasome in cerebral ischemia will be specified, as well as their relation to well-known pathways activating the innate immune response (e.g., Toll-like receptor signaling) and the consequences that result from their activation (beneficial versus deleterious).
Collapse
|
73
|
Obrenovitch TP. Molecular physiology of preconditioning-induced brain tolerance to ischemia. Physiol Rev 2008; 88:211-47. [PMID: 18195087 DOI: 10.1152/physrev.00039.2006] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Ischemic tolerance describes the adaptive biological response of cells and organs that is initiated by preconditioning (i.e., exposure to stressor of mild severity) and the associated period during which their resistance to ischemia is markedly increased. This topic is attracting much attention because preconditioning-induced ischemic tolerance is an effective experimental probe to understand how the brain protects itself. This review is focused on the molecular and related functional changes that are associated with, and may contribute to, brain ischemic tolerance. When the tolerant brain is subjected to ischemia, the resulting insult severity (i.e., residual blood flow, disruption of cellular transmembrane gradients) appears to be the same as in the naive brain, but the ensuing lesion is substantially reduced. This suggests that the adaptive changes in the tolerant brain may be primarily directed against postischemic and delayed processes that contribute to ischemic damage, but adaptive changes that are beneficial during the subsequent test insult cannot be ruled out. It has become clear that multiple effectors contribute to ischemic tolerance, including: 1) activation of fundamental cellular defense mechanisms such as antioxidant systems, heat shock proteins, and cell death/survival determinants; 2) responses at tissue level, especially reduced inflammatory responsiveness; and 3) a shift of the neuronal excitatory/inhibitory balance toward inhibition. Accordingly, an improved knowledge of preconditioning/ischemic tolerance should help us to identify neuroprotective strategies that are similar in nature to combination therapy, hence potentially capable of suppressing the multiple, parallel pathophysiological events that cause ischemic brain damage.
Collapse
Affiliation(s)
- Tihomir Paul Obrenovitch
- Division of Pharmacology, School of Life Sciences, University of Bradford, Bradford, United Kingdom.
| |
Collapse
|
74
|
Shaftel SS, Griffin WST, O'Banion MK. The role of interleukin-1 in neuroinflammation and Alzheimer disease: an evolving perspective. J Neuroinflammation 2008; 5:7. [PMID: 18302763 PMCID: PMC2335091 DOI: 10.1186/1742-2094-5-7] [Citation(s) in RCA: 358] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Accepted: 02/26/2008] [Indexed: 12/15/2022] Open
Abstract
Elevation of the proinflammatory cytokine Interleukin-1 (IL-1) is an integral part of the local tissue reaction to central nervous system (CNS) insult. The discovery of increased IL-1 levels in patients following acute injury and in chronic neurodegenerative disease laid the foundation for two decades of research that has provided important details regarding IL-1's biology and function in the CNS. IL-1 elevation is now recognized as a critical component of the brain's patterned response to insults, termed neuroinflammation, and of leukocyte recruitment to the CNS. These processes are believed to underlie IL-1's function in the setting of acute brain injury, where it has been ascribed potential roles in repair as well as in exacerbation of damage. Explorations of IL-1's role in chronic neurodegenerative disease have mainly focused on Alzheimer disease (AD), where indirect evidence has implicated it in disease pathogenesis. However, recent observations in animal models challenge earlier assumptions that IL-1 elevation and resulting neuroinflammatory processes play a purely detrimental role in AD, and prompt a need for new characterizations of IL-1 function. Potentially adaptive functions of IL-1 elevation in AD warrant further mechanistic studies, and provide evidence that enhancement of these effects may help to alleviate the pathologic burden of disease.
Collapse
Affiliation(s)
- Solomon S Shaftel
- Department of Neurobiology and Anatomy, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.
| | | | | |
Collapse
|
75
|
Clark SR, McMahon CJ, Gueorguieva I, Rowland M, Scarth S, Georgiou R, Tyrrell PJ, Hopkins SJ, Rothwell NJ. Interleukin-1 receptor antagonist penetrates human brain at experimentally therapeutic concentrations. J Cereb Blood Flow Metab 2008; 28:387-94. [PMID: 17684519 DOI: 10.1038/sj.jcbfm.9600537] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The proinflammatory cytokine interleukin (IL)-1 mediates several forms of experimentally induced acute brain injury and has been implicated in chronic neurodegenerative disorders. The IL-1 receptor antagonist, IL-1RA, protects rodents against ischaemic brain injury, but its molecular mass (17 kDa) potentially limits the brain penetration of peripherally administered IL-1RA. We therefore sought to identify whether therapeutically effective concentrations of IL-1RA in the rat were also achieved in brain of patients with subarachnoid haemorrhage (SAH), using a peripheral administration regime that had proved to be safe and reduce peripheral inflammation in patients after stroke. An intravenous bolus of IL-1RA, followed by infusion, was administered to rats after induction of focal cerebral ischaemia. The effects of IL-1RA on brain ischaemia and the concentrations achieved in cerebrospinal fluid (CSF), were determined. Interleukin-1 receptor antagonist was similarly administered to patients with SAH, and CSF was sampled via external ventricular drains. In rats, IL-1RA significantly reduced brain injury induced by focal cerebral ischaemia. The plasma IL-1RA concentrations reached 12+/-2 microg/mL by 30 mins, and CSF concentrations were maintained between 91 and 232 ng/mL between 1 and 24 h of infusion. In patients with SAH, IL-1RA reached a steady-state plasma concentration of 22+/-4 microg/mL by 15 mins, and CSF concentrations were maintained at 78 to 558 ng/mL between 1 and 24 h. Intravenous delivery of IL-1RA leads to CSF concentrations in patients comparable to those that are neuroprotective in rats, and might therefore be of therapeutic benefit.
Collapse
|
76
|
Nakka VP, Gusain A, Mehta SL, Raghubir R. Molecular mechanisms of apoptosis in cerebral ischemia: multiple neuroprotective opportunities. Mol Neurobiol 2008; 37:7-38. [PMID: 18066503 DOI: 10.1007/s12035-007-8013-9] [Citation(s) in RCA: 210] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2007] [Accepted: 11/05/2007] [Indexed: 12/18/2022]
Abstract
Cerebral ischemia/reperfusion (I/R) injury triggers multiple and distinct but overlapping cell signaling pathways, which may lead to cell survival or cell damage. There is overwhelming evidence to suggest that besides necrosis, apoptosis do contributes significantly to the cell death subsequent to I/R injury. Both extrinsic and intrinsic apoptotic pathways play a vital role, and upon initiation, these pathways recruit downstream apoptotic molecules to execute cell death. Caspases and Bcl-2 family members appear to be crucial in regulating multiple apoptotic cell death pathways initiated during I/R. Similarly, inhibitor of apoptosis family of proteins (IAPs), mitogen-activated protein kinases, and newly identified apoptogenic molecules, like second mitochondrial-activated factor/direct IAP-binding protein with low pI (Smac/Diablo), omi/high-temperature requirement serine protease A2 (Omi/HtrA2), X-linked mammalian inhibitor of apoptosis protein-associated factor 1, and apoptosis-inducing factor, have emerged as potent regulators of cellular apoptotic/antiapoptotic machinery. All instances of cell survival/death mechanisms triggered during I/R are multifaceted and interlinked, which ultimately decide the fate of brain cells. Moreover, apoptotic cross-talk between major subcellular organelles suggests that therapeutic strategies should be optimally directed at multiple targets/mechanisms for better therapeutic outcome. Based on the current knowledge, this review briefly focuses I/R injury-induced multiple mechanisms of apoptosis, involving key apoptotic regulators and their emerging roles in orchestrating cell death programme. In addition, we have also highlighted the role of autophagy in modulating cell survival/death during cerebral ischemia. Furthermore, an attempt has been made to provide an encouraging outlook on emerging therapeutic approaches for cerebral ischemia.
Collapse
Affiliation(s)
- Venkata Prasuja Nakka
- Division of Pharmacology, Central Drug Research Institute, Chatter Manzil Palace, POB-173, Lucknow, 226001, India
| | | | | | | |
Collapse
|
77
|
Harry GJ, Funk JA, Lefebvre d'Hellencourt C, McPherson CA, Aoyama M. The type 1 interleukin 1 receptor is not required for the death of murine hippocampal dentate granule cells and microglia activation. Brain Res 2007; 1194:8-20. [PMID: 18191113 DOI: 10.1016/j.brainres.2007.11.076] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Revised: 11/18/2007] [Accepted: 11/26/2007] [Indexed: 02/06/2023]
Abstract
Alterations in inflammatory process, neuronal death, and glia response have been observed under manipulation of interleukin-1 (IL-1) and subsequent signaling through the type 1 IL-1 receptor (IL-1R1). To investigate the influence of IL-1R1 activation in the pathophysiology of a chemical-induced injury to the murine hippocampus, we examined the level and pattern of neuronal death and neuroinflammation in male weanling mice exposed to trimethyltin hydroxide (2.0 mg TMT/kg, i.p.). Dentate granule cell death occurred at 6 h post-TMT as detected by active caspase 3 immunostaining and presence of lectin positive microglia. The severity of neuronal death and microglia response increased by 12-24 h with elevations in mRNA levels for TNFalpha and IL-1alpha. In IL-1R1 null (IL-1R1-/-) mice, the pattern and severity of neuronal death at 24 or 72 h post-TMT was similar as compared to wildtype (WT) mice. In both groups, mRNA levels for TNFalpha and MIP-1alpha were elevated, no significant change was seen in either IL-1alpha or IL-1beta, and the early activation of microglia, including their ability to progress to a phagocytic phenotype, was maintained. Compared to WT mice, IL-1R1-/- mice displayed a limited glial fibrillary acidic protein (GFAP) astrocytic response, as well as a preferential induction in mRNA levels of Fas signaling components. Cumulatively, these results indicate that IL-1R1 activation is not necessary for TMT-induced death of dentate granule neurons or local activation of microglia; however, IL-1R1 signaling is involved in mediating the structural response of astrocytes to injury and may regulate apoptotic mechanisms via Fas signaling components.
Collapse
Affiliation(s)
- G Jean Harry
- Neurotoxicology Group, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA.
| | | | | | | | | |
Collapse
|
78
|
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
|
79
|
Wolfberg AJ, Dammann O, Gressens P. Anti-inflammatory and immunomodulatory strategies to protect the perinatal brain. Semin Fetal Neonatal Med 2007; 12:296-302. [PMID: 17418653 DOI: 10.1016/j.siny.2007.01.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Infection and inflammation contribute to perinatal brain damage, particularly to the white matter. Although combating perinatal inflammation can be dangerous, because inflammation might have beneficial effects for mother and fetus, it is worthwhile reviewing potential anti-inflammatory neuroprotective compounds, along with their potential adverse effects. Further research on the possible neuroprotective roles of existing medications and substances is necessary.
Collapse
Affiliation(s)
- Adam J Wolfberg
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Tufts New England Medical Center, Boston, MA, USA
| | | | | |
Collapse
|
80
|
Cao L, Fei L, Chang TTL, DeLeo JA. Induction of interleukin-1beta by interleukin-4 in lipopolysaccharide-treated mixed glial cultures: microglial-dependent effects. J Neurochem 2007; 102:408-19. [PMID: 17442043 DOI: 10.1111/j.1471-4159.2007.04588.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Glial-secreted proinflammatory mediators are dynamically involved in central nervous system responses to exogenous stimuli such as infection, neurotoxins, and nerve injury. The therapeutic use of anti-inflammatory agents may reduce certain central nervous system pathology induced by inflammatory responses. We investigated the role of interleukin (IL)-4 in modulating the production of proinflammatory mediators from lipopolysaccharide-stimulated mixed glia in vitro. Interestingly, IL-4 significantly enhanced IL-1beta secretion and did not affect monocyte chemoattractant protein-1 release, even though IL-4 considerably inhibited IL-6, tumor necrosis factor alpha, and nitric oxide production from rat neonatal mixed glia. Further, IL-4 exhibited inhibitory effects on IL-1beta production in microglial-enriched cultures, while significantly increasing IL-1beta production in microglial-depleted glia. The enhancing effect of IL-4 on IL-1beta production was found to be inversely correlated with the percentage of microglia present in the mixed glial population. In summary, IL-4 did not act as a global anti-inflammatory cytokine and in fact, under certain situations enhanced IL-1beta secretion. We conclude that IL-4 exerts its anti-inflammatory effects in a limited and target-specific manner, which is delicately regulated by the cellular microenvironment. Therefore, precaution should be taken when clinically using IL-4 to treat diseases manifested by overt inflammatory responses.
Collapse
Affiliation(s)
- Ling Cao
- Department of Anesthesiology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire 03756, USA
| | | | | | | |
Collapse
|
81
|
Wang Q, Tang XN, Yenari MA. The inflammatory response in stroke. J Neuroimmunol 2007; 184:53-68. [PMID: 17188755 PMCID: PMC1868538 DOI: 10.1016/j.jneuroim.2006.11.014] [Citation(s) in RCA: 898] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2006] [Accepted: 11/17/2006] [Indexed: 12/17/2022]
Abstract
Recent works in the area of stroke and brain ischemia has demonstrated the significance of the inflammatory response accompanying necrotic brain injury. Acutely, this response appears to contribute to ischemic pathology, and anti-inflammatory strategies have become popular. This chapter will discuss the current knowledge of the contribution of systemic and local inflammation in experimental stroke. It will review the role of specific cell types including leukocytes, endothelium, glia, microglia, the extracellular matrix and neurons. Intracellular inflammatory signaling pathways such as nuclear factor kappa beta and mitogen-activated protein kinases, and mediators produced by inflammatory cells such as cytokines, chemokines, reactive oxygen species and arachidonic acid metabolites will be reviewed as well as the potential for therapy in stroke and hypoxic-ischemic injury.
Collapse
Affiliation(s)
- Qing Wang
- Department of Neurology, University of California, San Francisco and the San Francisco Veterans Affairs Medical Center, San Francisco, CA
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA
| | - Xian Nan Tang
- Department of Neurology, University of California, San Francisco and the San Francisco Veterans Affairs Medical Center, San Francisco, CA
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA
| | - Midori A. Yenari
- Department of Neurology, University of California, San Francisco and the San Francisco Veterans Affairs Medical Center, San Francisco, CA
| |
Collapse
|
82
|
Imai F, Suzuki H, Oda J, Ninomiya T, Ono K, Sano H, Sawada M. Neuroprotective effect of exogenous microglia in global brain ischemia. J Cereb Blood Flow Metab 2007; 27:488-500. [PMID: 16820801 DOI: 10.1038/sj.jcbfm.9600362] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Exogenous microglia pass through the blood-brain barrier and migrate to ischemic hippocampal lesions when injected into the circulation. We investigated the effect of exogenous microglia on ischemic CA1 pyramidal neurons. Microglia were isolated from neonatal mixed brain cultures, labeled with the fluorescent dye PKH26, and injected into the subclavian artery of Mongolian gerbils subjected to ischemia reperfusion neuronal injury. PKH26-labeled microglia migrated to the ischemic hippocampal lesion, resulting in increased numbers of surviving neurons compared with control animals, even when injected 24 h after ischemia. Interferon-gamma stimulation of isolated microglia enhanced the neuroprotective effect. Administration of exogenous microglia resulted in normal performance in a passive avoidance-learning task. Additionally, administration of exogenous microglia increased the expression of brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor in the ischemic hippocampus, and thus might have induced neurotrophin-dependent protective activity in damaged neurons. Peripherally injected microglia exhibited a specific affinity for ischemic brain lesions, and protected against ischemic neuronal injury in vivo. It is possible that administration of exogenous microglia can be developed as a potential candidate therapy for central nervous system repair after transitory global ischemia.
Collapse
Affiliation(s)
- Fumihiro Imai
- Department of Neurosurgery, Fujita Health University, Toyoake, Aichi, Japan.
| | | | | | | | | | | | | |
Collapse
|
83
|
Boutin H, Chauveau F, Thominiaux C, Kuhnast B, Grégoire MC, Jan S, Trebossen R, Dollé F, Tavitian B, Mattner F, Katsifis A. In vivo imaging of brain lesions with [11C]CLINME, a new PET radioligand of peripheral benzodiazepine receptors. Glia 2007; 55:1459-68. [PMID: 17680643 DOI: 10.1002/glia.20562] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The peripheral benzodiazepine receptor (PBR) is expressed by microglial cells in many neuropathologies involving neuroinflammation. PK11195, the reference compound for PBR, is used for positron emission tomography (PET) imaging but has a limited capacity to quantify PBR expression. Here we describe the new PBR ligand CLINME as an alternative to PK11195. In vitro and in vivo imaging properties of [(11)C]CLINME were studied in a rat model of local acute neuroinflammation, and compared with the reference compound [(11)C]PK11195, using autoradiography and PET imaging. Immunohistochemistry study was performed to validate the imaging data. [(11)C]CLINME exhibited a higher contrast between the PBR-expressing lesion site and the intact side of the same rat brain than [(11)C]PK11195 (2.14 +/- 0.09 vs. 1.62 +/- 0.05 fold increase, respectively). The difference was due to a lower uptake for [(11)C]CLINME than for [(11)C]PK11195 in the non-inflammatory part of the brain in which PBR was not expressed, while uptake levels in the lesion were similar for both tracers. Tracer localization correlated well with that of activated microglial cells, demonstrated by immunohistochemistry and PBR expression detected by autoradiography. Modeling using the simplified tissue reference model showed that R(1) was similar for both ligands (R(1) approximately 1), with [(11)C]CLINME exhibiting a higher binding potential than [(11)C]PK11195 (1.07 +/- 0.30 vs. 0.66 +/- 0.15). The results show that [(11)C]CLINME performs better than [(11)C]PK11195 in this model. Further studies of this new compound should be carried out to better define its capacity to overcome the limitations of [(11)C]PK11195 for PBR PET imaging.
Collapse
Affiliation(s)
- Hervé Boutin
- CEA, DSV, I2BM, SHFJ, Laboratoire d'Imagerie Moléculaire Expérimentale, Orsay, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Sébire G, Larouche A, Girard S, Roy M, Kadhim H. Effets pervers des cytokines sur le développement cérébral périnatal ? Med Sci (Paris) 2006; 22:1015-6. [PMID: 17156714 DOI: 10.1051/medsci/200622121015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
85
|
Selvarajah JR, Parry-Jones A, McMahon CJ, Rothwell NJ. Interleukin-1 as a therapeutic target in acute brain injury. FUTURE NEUROLOGY 2006. [DOI: 10.2217/14796708.1.4.417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Interest in the interactions between the immune and central nervous systems has furthered our understanding of brain function in health and disease. Experimental and clinical studies increasingly reveal an inflammatory component in the pathophysiology of many forms of brain injury. Members of the interleukin (IL)-1 cytokine family are produced by diverse cell types within the brain and may determine the outcome of neuronal injury. This review discusses the neuroprotective potential of IL-1 inhibition in various acute neurological and neurosurgical diseases. The ILs comprise an expanding family of cytokines with diverse physiological and pathological actions, of which IL-1 is a key inflammatory mediator implicated in brain injury. IL-1 expression in the normal CNS is low, but is upregulated rapidly in response to brain injury. In experimental studies, IL-1 and its endogenous, competitive, selective antagonist, IL-1 receptor antagonist (IL-1RA), mediate neurotoxic and neuroprotective outcomes, respectively. Clinical studies support the relationship between inflammation, disease severity and poor prognosis in various neurological and neurosurgical disorders. Recombinant human (rh)IL-1RA shows modest blood–brain barrier penetrance and is safe for clinical use when administered parenterally. Evidence supporting the use of rhIL-1RA and other approaches to targeting IL-1 are discussed in relation to cerebral ischemia, seizures, subarachnoid hemorrhage and traumatic brain injury.
Collapse
Affiliation(s)
- Johann R Selvarajah
- The University of Manchester, School of Medicine, Faculty of Medicine & Human Sciences, Manchester, UK
| | | | - Catherine J McMahon
- The University of Manchester, School of Medicine, Faculty of Medicine & Human Sciences, Manchester, UK
| | - Nancy J Rothwell
- The University of Manchester, Faculty of Life Sciences, Manchester, UK
| |
Collapse
|