Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
ACS Chem Biol ; 17(5): 1073-1081, 2022 05 20.
Article in English | MEDLINE | ID: mdl-35471821

ABSTRACT

We sought to develop a small-molecule activator of interferon regulatory factor 3 (IRF3), an essential innate immune transcription factor, which could potentially be used therapeutically in multiple disease settings. Using a high-throughput screen, we identified small-molecule entities that activate a type I interferon response, with minimal off-target NFκB activation. We identified 399 compounds at a hit rate of 0.24% from singlicate primary screening. Secondary screening included the primary hits and additional compounds with similar chemical structures obtained from other library sources and resulted in 142 candidate compounds. The hit compounds were sorted and ranked to identify compound groups with activity in both human and mouse backgrounds to facilitate animal model engagement for translational development. Chemical modifications within two groups of small molecules produced leads with improved activity over original hits. Furthermore, these leads demonstrated activity in ex vivo cytokine release assays from human blood- and mouse bone marrow-derived macrophages. Dependence on IRF3 was demonstrated using bone marrow-derived macrophages from IRF3-deficient mice, which were not responsive to the molecules. To identify the upstream pathway leading to IRF3 activation, we used a library of CRISPR knockout cell lines to test the key innate immune adaptor and receptor molecules. These studies indicated a surprising toll-interleukin-1 receptor-domain-containing-adapter-inducing interferon-ß-dependent but TLR3/4-independent mechanism of IRF3 activation.


Subject(s)
Interferon Regulatory Factor-3 , Signal Transduction , Animals , Antiviral Agents/pharmacology , Drug Development , Interferon Regulatory Factor-3/metabolism , Macrophages/metabolism , Mice
2.
Anal Bioanal Chem ; 413(10): 2735-2745, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33078250

ABSTRACT

Ischemic stroke is one of the major causes of death and permanent disability in the world. However, the molecular mechanisms surrounding tissue damage are complex and further studies are needed to gain insights necessary for development of treatment. Prophylactic treatment by administration of cytosine-guanine (CpG) oligodeoxynucleotides has been shown to provide neuroprotection against anticipated ischemic injury. CpG binds to Toll-like receptor 9 (TLR9) causing initialization of an inflammatory response that limits visible ischemic damages upon subsequent stroke. Here, we use nanospray desorption electrospray ionization (nano-DESI) mass spectrometry imaging (MSI) to characterize molecular effects of CpG preconditioning prior to middle cerebral artery occlusion (MCAO) and reperfusion. By doping the nano-DESI solvent with appropriate internal standards, we can study and compare distributions of phosphatidylcholine (PC) and lysophosphatidylcholine (LPC) in the ischemic hemisphere of the brain despite the large changes in alkali metal abundances. Our results show that CpG preconditioning not only reduces the infarct size but it also decreases the degradation of PC and accumulation of LPC species, which indicates reduced cell membrane breakdown and overall ischemic damage. Our findings show that molecular mechanisms of PC degradation are intact despite CpG preconditioning but that these are limited due to the initialized inflammatory response.


Subject(s)
Brain Chemistry , Brain/pathology , Infarction, Middle Cerebral Artery/therapy , Lysophosphatidylcholines/analysis , Oligodeoxyribonucleotides/therapeutic use , Animals , Disease Models, Animal , Infarction, Middle Cerebral Artery/pathology , Male , Mass Spectrometry , Mice, Inbred C57BL , Oligodeoxyribonucleotides/administration & dosage
3.
Transl Stroke Res ; 10(4): 440-448, 2019 08.
Article in English | MEDLINE | ID: mdl-30341719

ABSTRACT

Each year, thousands of patients are at risk of cerebral ischemic injury, due to iatrogenic responses to surgical procedures. Prophylactic treatment of these patients as standard care could minimize potential neurological complications. We have shown that protection of brain tissue, in a non-human primate model of cerebral ischemic injury, is possible through pharmacological preconditioning using the immune activator D192935. We postulate that preconditioning with D192935 results in neuroprotective reprogramming that is evident in the brain following experimentally induced cerebral ischemia. We performed quantitative proteomic analysis of cerebral spinal fluid (CSF) collected post-stroke from our previously published efficacy study to determine whether CSF protein profiles correlated with induced protection. Four groups of animals were examined: naïve animals (no treatment or stroke); animals treated with vehicle prior to stroke; D192935 treated and stroked animals, further delineated into two groups, ones that were protected (small infarcts) and those that were not protected (large infarcts). We found that distinct protein clusters defined the protected and non-protected animal groups, with a 16-member cluster of proteins induced exclusively in D192935 protected animals. Seventy percent of the proteins induced in the protected animals have functions that would enhance neuroprotection and tissue repair, including several members associated with M2 macrophages, a macrophage phenotype shown to contribute to neuroprotection and repair during ischemic injury. These studies highlight the translational importance of CSF biomarkers in defining mechanism and monitoring responses to treatment in development of stroke therapeutics.


Subject(s)
Brain Ischemia/cerebrospinal fluid , Brain Ischemia/prevention & control , Ischemic Preconditioning/methods , Neuroprotection/physiology , Proteomics/methods , Animals , Brain Ischemia/pathology , Macaca mulatta , Male , Neuroprotection/drug effects , Toll-Like Receptor 9/agonists
4.
Transl Stroke Res ; 8(4): 322-333, 2017 08.
Article in English | MEDLINE | ID: mdl-28378315

ABSTRACT

Ischemia-reperfusion brain injury can be iatrogenically induced secondary to life-saving procedures. Prophylactic treatment of these patients offers a promising prevention for lifelong complications. We postulate that a cytosine-guanine (CpG) oligodeoxynucleotide (ODN) can provide robust antecedent protection against cerebral ischemic injury with minimal release of pro-inflammatory cytokines, making it an ideal candidate for further clinical development. Mouse and nonhuman primate (NHP) models of cerebral ischemic injury were used to test whether an A-type CpG ODN, which induces minimal systemic inflammatory cytokine responses, can provide prophylactic protection. Extent of injury in the mouse was measured by histological staining of live tissue. In the NHP, injury was assessed 2 and 7 days post-occlusion from T2-weighted magnetic resonance images and neurological and motor deficits were cataloged daily. Plasma cytokine levels were measured using species-specific Luminex assays. Prophylactic administration of an A-type CpG ODN provided robust protection against cerebral ischemic injury in the mouse with minimal systemic inflammation. Rhesus macaques treated with D192935, a mixture of human optimized A-type CpG ODNs, had smaller infarcts and demonstrated significantly less neurological and motor deficits following ischemic injury. Our findings demonstrate the translational potential of D192935 as a prophylactic treatment for patients at risk of cerebral ischemic injury.


Subject(s)
Brain Ischemia/prevention & control , Cytokines/metabolism , Drug Evaluation, Preclinical , Neuroprotective Agents/therapeutic use , Oligodeoxyribonucleotides/therapeutic use , Reperfusion Injury/prevention & control , Analysis of Variance , Animals , Brain Infarction/diagnostic imaging , Brain Infarction/drug therapy , Brain Infarction/etiology , Disease Models, Animal , Dose-Response Relationship, Drug , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Neurologic Examination , Neuroprotective Agents/chemistry , Oligodeoxyribonucleotides/chemistry , Physical Conditioning, Animal/physiology , Reperfusion Injury/complications , Time Factors
5.
Stroke ; 47(1): 262-6, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26564103

ABSTRACT

BACKGROUND AND PURPOSE: Preconditioning with poly-l-lysine and carboxymethylcellulose (ICLC) provides robust neuroprotection from cerebral ischemia in a mouse stroke model. However, the receptor that mediates neuroprotection is unknown. As a synthetic double-stranded RNA, poly-ICLC may bind endosomal Toll-like receptor 3 or one of the cytosolic retinoic acid-inducible gene-I-like receptor family members, retinoic acid-inducible gene-I, or melanoma differentiation-associated protein 5. Activation of these receptors culminates in type I interferons (IFN-α/ß) induction-a response required for poly-ICLC-induced neuroprotection. In this study, we investigate the receptor required for poly-ICLC-induced neuroprotection. METHODS: Toll-like receptor 3, melanoma differentiation-associated protein 5-, and IFN-promoter stimulator 1-deficient mice were treated with poly-ICLC 24 hours before middle cerebral artery occlusion. Infarct volume was measured 24 hours after stroke to identify the receptor signaling pathways involved in protection. IFN-α/ß induction was measured in plasma samples collected 6 hours after poly-ICLC treatment. IFN-ß-deficient mice were used to test the requirement of IFN-ß for poly-ICLC-induced neuroprotection. Mice were treated with recombinant IFN-α-A to test the role of IFN-α as a potential mediator of neuroprotection. RESULTS: Poly-ICLC induction of both neuroprotection and systemic IFN-α/ß requires the cytosolic receptor melanoma differentiation-associated protein 5 and the adapter molecule IFN-promoter stimulator 1, whereas it is independent of Toll-like receptor 3. IFN-ß is not required for poly-ICLC-induced neuroprotection. IFN-α treatment protects against stroke. CONCLUSIONS: Poly-ICLC preconditioning is mediated by melanoma differentiation-associated protein 5 and its adaptor molecule IFN-promoter stimulator 1. This is the first evidence that a cytosolic receptor can mediate neuroprotection, providing a new target for the development of therapeutic agents to protect the brain from ischemic injury.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/prevention & control , DEAD-box RNA Helicases/metabolism , Ischemic Preconditioning/methods , Stroke/metabolism , Stroke/prevention & control , Animals , Carboxymethylcellulose Sodium/analogs & derivatives , Carboxymethylcellulose Sodium/metabolism , Carboxymethylcellulose Sodium/therapeutic use , Interferon-Induced Helicase, IFIH1 , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroprotective Agents/metabolism , Neuroprotective Agents/therapeutic use , Poly I-C/metabolism , Poly I-C/therapeutic use , Polylysine/analogs & derivatives , Polylysine/metabolism , Polylysine/therapeutic use
6.
Acta Neurochir Suppl ; 121: 39-44, 2016.
Article in English | MEDLINE | ID: mdl-26463920

ABSTRACT

Stroke activates an inflammatory response that results in the infiltration of peripheral immune cells into the ischemic area, contributing to exacerbation of tissue damage. However, evidence indicates that inflammatory cell infiltration can also promote neuroprotection through regulatory immune cells that mitigate injury. These immune regulatory cells may also be important mediators of neuroprotection associated with preconditioning, a phenomenon whereby small exposure to a potential harmful stimulus is able to induce protection against a subsequent ischemic event. The elucidation of mechanisms that allow these immune cells to confer neuroprotection is critical to developing new therapeutic strategies against acute stroke. In the present review, we discuss the dual role of peripheral immune cells in stroke-related brain injury and neuroprotection. Furthermore, we report new data from our laboratory that supports the important role of peripheral cells and their interaction with the brain endothelium for the establishment of the protective phenotype in preconditioning.


Subject(s)
Ischemic Preconditioning , Lymphocytes/immunology , Macrophages/immunology , Neuroprotection/immunology , Neutrophils/immunology , Stroke/immunology , Animals , Humans
7.
J Cereb Blood Flow Metab ; 35(2): 257-66, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25388675

ABSTRACT

Cytosine-phosphate-guanine (CpG) preconditioning reprograms the genomic response to stroke to protect the brain against ischemic injury. The mechanisms underlying genomic reprogramming are incompletely understood. MicroRNAs (miRNAs) regulate gene expression; however, their role in modulating gene responses produced by CpG preconditioning is unknown. We evaluated brain miRNA expression in response to CpG preconditioning before and after stroke using microarray. Importantly, we have data from previous gene microarrays under the same conditions, which allowed integration of miRNA and gene expression data to specifically identify regulated miRNA gene targets. CpG preconditioning did not significantly alter miRNA expression before stroke, indicating that miRNA regulation is not critical for the initiation of preconditioning-induced neuroprotection. However, after stroke, differentially regulated miRNAs between CpG- and saline-treated animals associated with the upregulation of several neuroprotective genes, implicating these miRNAs in genomic reprogramming that increases neuroprotection. Statistical analysis revealed that the miRNA targets were enriched in the gene population regulated in the setting of stroke, implying that miRNAs likely orchestrate this gene expression. These data suggest that miRNAs regulate endogenous responses to stroke and that manipulation of these miRNAs may have the potential to acutely activate novel neuroprotective processes that reduce damage.


Subject(s)
Brain Ischemia/drug therapy , Gene Expression Regulation/drug effects , MicroRNAs/biosynthesis , Neuroprotective Agents/pharmacology , Oligodeoxyribonucleotides/pharmacology , Stroke/drug therapy , Adjuvants, Immunologic/pharmacology , Animals , Brain Ischemia/metabolism , Male , Mice , Oligonucleotide Array Sequence Analysis , Stroke/metabolism , Stroke/pathology
9.
Analyst ; 139(14): 3528-32, 2014 Jul 21.
Article in English | MEDLINE | ID: mdl-24802717

ABSTRACT

Matrix effects in mass spectrometry imaging (MSI) may affect the observed molecular distribution in chemical and biological systems. In this study, we use mouse brain tissue of a middle cerebral artery occlusion (MCAO) stroke model to examine matrix effects in nanospray desorption electrospray ionization MSI (nano-DESI MSI). This is achieved by normalizing the intensity of the sodium and potassium adducts of endogenous phosphatidylcholine (PC) species to the intensity of the corresponding adduct of the PC standard supplied at a constant rate with the nano-DESI solvent. The use of MCAO model with an ischemic region localized to one hemisphere of the brain enables immediate comparison of matrix effects within one ion image. Furthermore, significant differences in sodium and potassium concentrations in the ischemic region in comparison with the healthy tissue allowed us to distinguish between two types of matrix effects. Specifically, we discuss matrix effects originating from variations in alkali metal concentrations and matrix effects originating from variations in the molecular composition of the tissue. Compensation for both types of matrix effects was achieved by normalizing the signals corresponding to endogenous PC to the signals of the standards. This approach, which does not introduce any complexity in sample preparation, efficiently compensates for signal variations resulting from differences in the local concentrations of sodium and potassium in tissue sections and from the complexity of the extracted analyte mixture derived from local variations in molecular composition.


Subject(s)
Brain Chemistry , Brain/blood supply , Brain/pathology , Infarction, Middle Cerebral Artery/pathology , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Infarction, Middle Cerebral Artery/diagnosis , Mice , Mice, Inbred C57BL , Optical Imaging/methods , Phosphatidylcholines/analysis , Potassium/analysis , Sodium/analysis
10.
PLoS Comput Biol ; 8(10): e1002722, 2012.
Article in English | MEDLINE | ID: mdl-23071432

ABSTRACT

The ability to examine the behavior of biological systems in silico has the potential to greatly accelerate the pace of discovery in diseases, such as stroke, where in vivo analysis is time intensive and costly. In this paper we describe an approach for in silico examination of responses of the blood transcriptome to neuroprotective agents and subsequent stroke through the development of dynamic models of the regulatory processes observed in the experimental gene expression data. First, we identified functional gene clusters from these data. Next, we derived ordinary differential equations (ODEs) from the data relating these functional clusters to each other in terms of their regulatory influence on one another. Dynamic models were developed by coupling these ODEs into a model that simulates the expression of regulated functional clusters. By changing the magnitude of gene expression in the initial input state it was possible to assess the behavior of the networks through time under varying conditions since the dynamic model only requires an initial starting state, and does not require measurement of regulatory influences at each time point in order to make accurate predictions. We discuss the implications of our models on neuroprotection in stroke, explore the limitations of the approach, and report that an optimized dynamic model can provide accurate predictions of overall system behavior under several different neuroprotective paradigms.


Subject(s)
Gene Regulatory Networks , Models, Genetic , Stroke/genetics , Stroke/metabolism , Transcriptome , Algorithms , Animals , Brain Ischemia/genetics , Brain Ischemia/metabolism , Computer Simulation , Gene Expression Profiling , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Multigene Family , Reproducibility of Results
11.
J Neurochem ; 123 Suppl 2: 75-85, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23050645

ABSTRACT

Preconditioning with a low dose of harmful stimulus prior to injury induces tolerance to a subsequent ischemic challenge resulting in neuroprotection against stroke. Experimental models of preconditioning primarily focus on neurons as the cellular target of cerebral protection, while less attention has been paid to the cerebrovascular compartment, whose role in the pathogenesis of ischemic brain injury is crucial. We have shown that preconditioning with polyinosinic polycytidylic acid (poly-ICLC) protects against cerebral ischemic damage. To delineate the mechanism of poly-ICLC protection, we investigated whether poly-ICLC preconditioning preserves the function of the blood-brain barrier (BBB) in response to ischemic injury. Using an in vitro BBB model, we found that poly-ICLC treatment prior to exposure to oxygen-glucose deprivation maintained the paracellular and transcellular transport across the endothelium and attenuated the drop in transendothelial electric resistance. We found that poly-ICLC treatment induced interferon (IFN) ß mRNA expression in astrocytes and microglia and that type I IFN signaling in brain microvascular endothelial cells was required for protection. Importantly, this implicates a potential mechanism underlying neuroprotection in our in vivo experimental stroke model, where type I IFN signaling is required for poly-ICLC-induced neuroprotection against ischemic injury. In conclusion, we are the first to show that preconditioning with poly-ICLC attenuates ischemia-induced BBB dysfunction. This mechanism is likely an important feature of poly-ICLC-mediated neuroprotection and highlights the therapeutic potential of targeting BBB signaling pathways to protect the brain against stroke.


Subject(s)
Blood-Brain Barrier/drug effects , Carboxymethylcellulose Sodium/analogs & derivatives , Infarction, Middle Cerebral Artery/prevention & control , Interferon Regulatory Factor-1/metabolism , Ischemic Preconditioning/methods , Neuroprotective Agents/administration & dosage , Poly I-C/administration & dosage , Polylysine/analogs & derivatives , Signal Transduction/drug effects , Analysis of Variance , Animals , Animals, Newborn , Blood-Brain Barrier/metabolism , Brain Infarction/drug therapy , Brain Infarction/etiology , Carboxymethylcellulose Sodium/administration & dosage , Carboxymethylcellulose Sodium/pharmacology , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glucose/deficiency , Hypoxia/drug therapy , Hypoxia/metabolism , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/genetics , Interferon Regulatory Factor-1/deficiency , Interferon-beta/genetics , Interferon-beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroglia/drug effects , Poly I-C/pharmacology , Polylysine/administration & dosage , Polylysine/pharmacology , RNA, Messenger/metabolism , Tight Junctions/drug effects , Tight Junctions/pathology , Time Factors
12.
J Cereb Blood Flow Metab ; 32(12): 2193-200, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23010947

ABSTRACT

Systemic preconditioning with the TLR9 ligand CpG induces neuroprotection against brain ischemic injury through a tumor necrosis factor (TNF)-dependent mechanism. It is unclear how systemic administration of CpG engages the brain to induce the protective phenotype. To address this, we created TLR9-deficient reciprocal bone marrow chimeric mice lacking TLR9 on either hematopoietic cells or radiation-resistant cells of nonhematopoietic origin. We report that wild-type mice reconstituted with TLR9-deficient hematopoietic cells failed to show neuroprotection after systemic CpG preconditioning. Further, while hematopoietic expression of TLR9 is required for CpG-induced neuroprotection it is not sufficient to restore protection to TLR9-deficient mice that are reconstituted with hematopoietic cells bearing TLR9. To determine whether the absence of protection was associated with TNF, we examined TNF levels in the systemic circulation and the brain. We found that although TNF is required for CpG preconditioning, systemic TNF levels did not correlate with the protective phenotype. However, induction of cerebral TNF mRNA required expression of TLR9 on both hematopoietic and nonhematopoietic cells and correlated with neuroprotection. In accordance with these results, we show the therapeutic potential of intranasal CpG preconditioning, which induces brain TNF mRNA and robust neuroprotection with no concomitant increase in systemic levels of TNF.


Subject(s)
Adjuvants, Immunologic/pharmacology , Bone Marrow Transplantation , Brain Ischemia/metabolism , Oligodeoxyribonucleotides/pharmacology , Toll-Like Receptor 9/biosynthesis , Transplantation Chimera/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Brain Injuries/genetics , Brain Injuries/metabolism , Brain Injuries/physiopathology , Brain Ischemia/genetics , Brain Ischemia/pathology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Mice , Mice, Knockout , Toll-Like Receptor 9/genetics , Transplantation Chimera/genetics , Transplantation, Homologous , Tumor Necrosis Factor-alpha/genetics
13.
PLoS One ; 7(6): e36465, 2012.
Article in English | MEDLINE | ID: mdl-22745654

ABSTRACT

The innate immune system plays important roles in a number of disparate processes. Foremost, innate immunity is a first responder to invasion by pathogens and triggers early defensive responses and recruits the adaptive immune system. The innate immune system also responds to endogenous damage signals that arise from tissue injury. Recently it has been found that innate immunity plays an important role in neuroprotection against ischemic stroke through the activation of the primary innate immune receptors, Toll-like receptors (TLRs). Using several large-scale transcriptomic data sets from mouse and mouse macrophage studies we identified targets predicted to be important in controlling innate immune processes initiated by TLR activation. Targets were identified as genes with high betweenness centrality, so-called bottlenecks, in networks inferred from statistical associations between gene expression patterns. A small set of putative bottlenecks were identified in each of the data sets investigated including interferon-stimulated genes (Ifit1, Ifi47, Tgtp and Oasl2) as well as genes uncharacterized in immune responses (Axud1 and Ppp1r15a). We further validated one of these targets, Ifit1, in mouse macrophages by showing that silencing it suppresses induction of predicted downstream genes by lipopolysaccharide (LPS)-mediated TLR4 activation through an unknown direct or indirect mechanism. Our study demonstrates the utility of network analysis for identification of interesting targets related to innate immune function, and highlights that Ifit1 can exert a positive regulatory effect on downstream genes.


Subject(s)
Carrier Proteins/metabolism , Immunity, Innate/immunology , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Cell Line , Immunity, Innate/drug effects , Immunity, Innate/genetics , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , RNA-Binding Proteins , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
14.
Stroke ; 43(5): 1383-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22403050

ABSTRACT

BACKGROUND AND PURPOSE: Systemic administration of Toll-like receptor (TLR) 4 and TLR9 agonists before cerebral ischemia have been shown to reduce ischemic injury by reprogramming the response of the brain to stroke. Our goal was to explore the mechanism of TLR-induced neuroprotection by determining whether a TLR7 agonist also protects against stroke injury. METHODS: C57Bl/6, TNF(-/-), interferon (IFN) regulatory factor 7(-/-), or type I IFN receptor (IFNAR)(-/-) mice were subcutaneously administered the TLR7 agonist Gardiquimod (GDQ) 72 hours before middle cerebral artery occlusion. Infarct volume and functional outcome were determined after reperfusion. Plasma cytokine responses and induction of mRNA for IFN-related genes in the brain were measured. IFNAR(-/-) mice also were treated with the TLR4 agonist (lipopolysaccharide) or the TLR9 agonist before middle cerebral artery occlusion and infarct volumes measured. RESULTS: The results show that GDQ reduces infarct volume as well as functional deficits in mice. GDQ pretreatment provided robust neuroprotection in TNF(-/-) mice, indicating that TNF was not essential. GDQ induced a significant increase in plasma IFNα levels and both IRF7(-/-) and IFNAR(-/-) mice failed to be protected, implicating a role for IFN signaling in TLR7-mediated protection. CONCLUSIONS: Our studies provide the first evidence that TLR7 preconditioning can mediate neuroprotection against ischemic injury. Moreover, we show that the mechanism of protection is unique from other TLR preconditioning ligands in that it is independent of TNF and dependent on IFNAR.


Subject(s)
Aminoquinolines/therapeutic use , Brain/blood supply , Imidazoles/therapeutic use , Ischemic Preconditioning/methods , Membrane Glycoproteins/agonists , Neuroprotective Agents/therapeutic use , Receptor, Interferon alpha-beta/physiology , Stroke/prevention & control , Toll-Like Receptor 7/agonists , Animals , Brain Infarction/pathology , Interferon Regulatory Factor-7/deficiency , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Signal Transduction/physiology , Stroke/physiopathology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/physiology
15.
J Cereb Blood Flow Metab ; 32(2): 242-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22086194

ABSTRACT

Preconditioning induces ischemic tolerance, which confers robust protection against ischemic damage. We show marked protection with polyinosinic polycytidylic acid (poly-IC) preconditioning in three models of murine ischemia-reperfusion injury. Poly-IC preconditioning induced protection against ischemia modeled in vitro in brain cortical cells and in vivo in models of brain ischemia and renal ischemia. Further, unlike other Toll-like receptor (TLR) ligands, which generally induce significant inflammatory responses, poly-IC elicits only modest systemic inflammation. Results show that poly-IC is a new powerful prophylactic treatment that offers promise as a clinical therapeutic strategy to minimize damage in patient populations at risk of ischemic injury.


Subject(s)
Brain Ischemia/prevention & control , Interferon Inducers/therapeutic use , Ischemic Preconditioning/methods , Kidney Diseases/prevention & control , Poly I-C/therapeutic use , Reperfusion Injury/prevention & control , Animals , Brain/drug effects , Brain/physiopathology , Brain Ischemia/physiopathology , Cells, Cultured , Cytokines/blood , Kidney/drug effects , Kidney/physiopathology , Kidney Diseases/physiopathology , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/therapeutic use , Reperfusion Injury/physiopathology
16.
J Neuroinflammation ; 8: 140, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21999375

ABSTRACT

BACKGROUND: Toll-like receptor 4 (TLR4) is activated in response to cerebral ischemia leading to substantial brain damage. In contrast, mild activation of TLR4 by preconditioning with low dose exposure to lipopolysaccharide (LPS) prior to cerebral ischemia dramatically improves outcome by reprogramming the signaling response to injury. This suggests that TLR4 signaling can be altered to induce an endogenously neuroprotective phenotype. However, the TLR4 signaling events involved in this neuroprotective response are poorly understood. Here we define several molecular mediators of the primary signaling cascades induced by LPS preconditioning that give rise to the reprogrammed response to cerebral ischemia and confer the neuroprotective phenotype. METHODS: C57BL6 mice were preconditioned with low dose LPS prior to transient middle cerebral artery occlusion (MCAO). Cortical tissue and blood were collected following MCAO. Microarray and qtPCR were performed to analyze gene expression associated with TLR4 signaling. EMSA and DNA binding ELISA were used to evaluate NFκB and IRF3 activity. Protein expression was determined using Western blot or ELISA. MyD88-/- and TRIF-/- mice were utilized to evaluate signaling in LPS preconditioning-induced neuroprotection. RESULTS: Gene expression analyses revealed that LPS preconditioning resulted in a marked upregulation of anti-inflammatory/type I IFN-associated genes following ischemia while pro-inflammatory genes induced following ischemia were present but not differentially modulated by LPS. Interestingly, although expression of pro-inflammatory genes was observed, there was decreased activity of NFκB p65 and increased presence of NFκB inhibitors, including Ship1, Tollip, and p105, in LPS-preconditioned mice following stroke. In contrast, IRF3 activity was enhanced in LPS-preconditioned mice following stroke. TRIF and MyD88 deficient mice revealed that neuroprotection induced by LPS depends on TLR4 signaling via TRIF, which activates IRF3, but does not depend on MyD88 signaling. CONCLUSION: Our results characterize several critical mediators of the TLR4 signaling events associated with neuroprotection. LPS preconditioning redirects TLR4 signaling in response to stroke through suppression of NFκB activity, enhanced IRF3 activity, and increased anti-inflammatory/type I IFN gene expression. Interestingly, this protective phenotype does not require the suppression of pro-inflammatory mediators. Furthermore, our results highlight a critical role for TRIF-IRF3 signaling as the governing mechanism in the neuroprotective response to stroke.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , Brain Ischemia , Interferon Regulatory Factor-3/immunology , Ischemic Preconditioning , Lipopolysaccharides/pharmacology , Signal Transduction , Stroke , Toll-Like Receptor 4/immunology , Adaptor Proteins, Vesicular Transport/genetics , Animals , Brain Ischemia/blood , Brain Ischemia/immunology , Brain Ischemia/pathology , Chemokines/blood , Chemokines/immunology , Cytokines/blood , Cytokines/immunology , Gene Expression Profiling , Humans , Infarction, Middle Cerebral Artery , Interferon Regulatory Factor-3/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , NF-kappa B/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Stroke/blood , Stroke/immunology , Stroke/pathology
17.
J Neurosci ; 31(23): 8456-63, 2011 Jun 08.
Article in English | MEDLINE | ID: mdl-21653850

ABSTRACT

Ischemic tolerance can be induced by numerous preconditioning stimuli, including various Toll-like receptor (TLR) ligands. We have shown previously that systemic administration of the TLR4 ligand LPS or the TLR9 ligand unmethylated CpG oligodeoxynucleotide before transient brain ischemia in mice confers substantial protection against ischemic damage. To elucidate the molecular mechanisms of preconditioning, we compared brain genomic profiles in response to preconditioning with these TLR ligands and with preconditioning via exposure to brief ischemia. We found that exposure to the TLR ligands and brief ischemia induced genomic changes in the brain characteristic of a TLR pathway-mediated response. Interestingly, all three preconditioning stimuli resulted in a reprogrammed response to stroke injury that converged on a shared subset of 13 genes not evident in the genomic profile from brains that were subjected to stroke without prior preconditioning. Analysis of the promoter region of these shared genes showed sequences required for interferon regulatory factor (IRF)-mediated transcription. The importance of this IRF gene network was tested using mice deficient in IRF3 or IRF7. Our data show that both transcription factors are required for TLR-mediated preconditioning and neuroprotection. These studies are the first to discover a convergent mechanism of neuroprotection induced by preconditioning--one that potentially results in reprogramming of the TLR-mediated response to stroke and requires the presence of IRF3 and IRF7.


Subject(s)
Brain Ischemia/immunology , Brain/immunology , Interferon Regulatory Factors/immunology , Ischemic Preconditioning/methods , Lipopolysaccharides/pharmacology , Toll-Like Receptors/immunology , Animals , Brain/blood supply , Brain/drug effects , Brain Ischemia/genetics , Gene Expression/drug effects , Interferon Regulatory Factors/genetics , Lipopolysaccharides/immunology , Male , Mice , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/immunology , Toll-Like Receptors/genetics
18.
Pac Symp Biocomput ; : 314-25, 2011.
Article in English | MEDLINE | ID: mdl-21121059

ABSTRACT

Determining biological network dependencies that can help predict the behavior of a system given prior observations from high-throughput data is a very valuable but difficult task, especially in the light of the ever-increasing volume of experimental data. Such an endeavor can be greatly enhanced by considering regulatory influences on co-expressed groups of genes representing functional modules, thus constraining the number of parameters in the system. This allows development of network models that are predictive of system dynamics. We first develop a predictive network model of the transcriptomics of whole blood from a mouse model of neuroprotection in ischemic stroke, and show that it can accurately predict system behavior under novel conditions. We then use a network topology approach to expand the set of regulators considered and show that addition of topological bottlenecks improves the performance of the predictive model. Finally, we explore how improvements in definition of functional modules may be achieved through an integration of inferred network relationships and functional relationships defined using Gene Ontology similarity. We show that appropriate integration of these two types of relationships can result in models with improved performance.


Subject(s)
Gene Expression Profiling/statistics & numerical data , Gene Regulatory Networks , Models, Genetic , Algorithms , Animals , Artificial Intelligence , Biomedical Engineering , Computational Biology , Disease Models, Animal , Mice , Oligonucleotide Array Sequence Analysis/statistics & numerical data , Stroke/genetics , Stroke/prevention & control , Systems Biology/statistics & numerical data
19.
J Neurosci ; 29(31): 9839-49, 2009 Aug 05.
Article in English | MEDLINE | ID: mdl-19657036

ABSTRACT

Lipopolysaccharide (LPS) preconditioning provides neuroprotection against subsequent cerebral ischemic injury through activation of its receptor, Toll-like receptor 4 (TLR4). Paradoxically, TLR activation by endogenous ligands after ischemia worsens stroke damage. Here, we define a novel, protective role for TLRs after ischemia in the context of LPS preconditioning. Microarray analysis of brains collected 24 h after stroke revealed a unique set of upregulated genes in LPS-pretreated animals. Promoter analysis of the unique gene set identified an overrepresentation of type I interferon (IFN)-associated transcriptional regulatory elements. This finding suggested the presence of type I IFNs or interferon regulatory factors (IRFs), which upregulate interferon-stimulated genes. Upregulation of IFNbeta was confirmed by real-time reverse transcription-PCR. Direct administration of IFNbeta intracerebroventricularly at the time of stroke was sufficient for neuroprotection. TLR4 can induce both IFNbeta and interferon-stimulated genes through its adapter molecule Toll/interleukin receptor domain-containing adaptor-inducing IFNbeta (TRIF) and the IRF3 transcription factor. We show in oxygen glucose deprivation of cortical neurons, an in vitro model of stroke, that activation of TRIF after stroke reduces neuronal death. Furthermore, mice lacking IRF3 were not protected by LPS preconditioning in our in vivo model. Our studies constitute the first demonstration of the neuroprotective capacity of TRIF/IRF3 signaling and suggest that interferon-stimulated genes, whether induced by IFNbeta or by enhanced TLR signaling to IRF3, are a potent means of protecting the brain against ischemic damage.


Subject(s)
Infarction, Middle Cerebral Artery/drug therapy , Interferon Regulatory Factor-3/metabolism , Interferon-beta/therapeutic use , Lipopolysaccharides/administration & dosage , Neuroprotective Agents/administration & dosage , Reperfusion Injury/prevention & control , Animals , Brain/drug effects , Brain/metabolism , Cell Death/drug effects , Cell Hypoxia/drug effects , Gene Expression/drug effects , Glucose/deficiency , Interferon Regulatory Factor-3/genetics , Interferon-beta/genetics , Interferon-beta/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/physiology , Oligonucleotide Array Sequence Analysis
20.
Stroke ; 40(3 Suppl): S34-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19064776

ABSTRACT

BACKGROUND AND PURPOSE: Systemic administration of cytosine-guanine (CpG) oligodeoxynucleotides provides neuroprotection against subsequent cerebral ischemic injury. We examined the genomic response of leukocytes and brain cells after ischemia in the context of CpG preconditioning. METHODS: RNA was isolated from circulating leukocytes and ischemic cortex 3 and 24 hours after middle cerebral artery occlusion after CpG or saline pretreatment and subjected to microarray analysis. Genes uniquely upregulated in CpG-pretreated mice were examined for overrepresented transcriptional regulatory elements. RESULTS: CpG preconditioning induced a novel response to middle cerebral artery occlusion within circulating leukocytes that was dominated by natural killer cell-associated genes and the GATA-3 transcriptional regulatory element. Preconditioning also caused a novel brain response to stroke that was dominated by Type I interferon, interferon-associated genes, and transcriptional regulatory elements. CONCLUSIONS: CpG preconditioning invokes novel leukocyte and brain responses to stroke. In this, CpG may be a unique preconditioning agent, coordinating peripheral and brain responses to protect against ischemic injury.


Subject(s)
Brain Ischemia/prevention & control , Brain Ischemia/physiopathology , Encephalitis/physiopathology , Ischemic Preconditioning , Toll-Like Receptors/physiology , Acute Disease , Animals , Arterial Occlusive Diseases/pathology , Arterial Occlusive Diseases/physiopathology , Arterial Occlusive Diseases/prevention & control , Brain Ischemia/pathology , Cerebral Arteries/metabolism , Cerebral Arteries/pathology , Cerebral Arteries/physiopathology , Dinucleoside Phosphates/pharmacology , Disease Models, Animal , Encephalitis/metabolism , Encephalitis/pathology , GATA3 Transcription Factor/metabolism , Interferon Type I/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Male , Mice , Mice, Inbred C57BL , Stroke/pathology , Stroke/physiopathology , Stroke/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL