Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Mol Ther Nucleic Acids ; 19: 1290-1298, 2020 Mar 06.
Article in English | MEDLINE | ID: mdl-32092825

ABSTRACT

Hundreds of dominant-negative myosin mutations have been identified that lead to hypertrophic cardiomyopathy, and the biomechanical link between mutation and disease is heterogeneous across this patient population. To increase the therapeutic feasibility of treating this diverse genetic population, we investigated the ability of locked nucleic acid (LNA)-modified antisense oligonucleotides (ASOs) to selectively knock down mutant myosin transcripts by targeting single-nucleotide polymorphisms (SNPs) that were found to be common in the myosin heavy chain 7 (MYH7) gene. We identified three SNPs in MYH7 and designed ASO libraries to selectively target either the reference or alternate MYH7 sequence. We identified ASOs that selectively knocked down either the reference or alternate allele at all three SNP regions. We also show allele-selective knockdown in a mouse model that was humanized on one allele. These results suggest that SNP-targeting ASOs are a promising therapeutic modality for treating cardiac pathology.

2.
Nucleic Acid Ther ; 23(5): 302-10, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23952551

ABSTRACT

Antisense oligonucleotides that recruit RNase H and thereby cleave complementary messenger RNAs are being developed as therapeutics. Dose-dependent hepatic changes associated with hepatocyte necrosis and increases in serum alanine-aminotransferase levels have been observed after treatment with certain oligonucleotides. Although general mechanisms for drug-induced hepatic injury are known, the characteristics of oligonucleotides that determine their hepatotoxic potential are not well understood. Here, we present a comprehensive analysis of the hepatotoxic potential of locked nucleic acid-modified oligonucleotides in mice. We developed a random forests classifier, in which oligonucleotides are regarded as being composed of dinucleotide units, which distinguished between 206 oligonucleotides with high and low hepatotoxic potential with 80% accuracy as estimated by out-of-bag validation. In a validation set, 17 out of 23 oligonucleotides were correctly predicted (74% accuracy). In isolation, some dinucleotide units increase, and others decrease, the hepatotoxic potential of the oligonucleotides within which they are found. However, a complex interplay between all parts of an oligonucleotide can influence the hepatotoxic potential. Using the classifier, we demonstrate how an oligonucleotide with otherwise high hepatotoxic potential can be efficiently redesigned to abate hepatotoxic potential. These insights establish analysis of sequence and modification patterns as a powerful tool in the preclinical discovery process for oligonucleotide-based medicines.


Subject(s)
Alanine Transaminase/blood , Drug Design , Liver/drug effects , Oligonucleotides, Antisense/toxicity , Oligonucleotides/toxicity , Phosphorothioate Oligonucleotides/toxicity , Algorithms , Animals , Body Weight , Female , Liver/pathology , Mice , Mice, Inbred C57BL , Nucleic Acid Conformation , Oligonucleotides/administration & dosage , Oligonucleotides/chemical synthesis , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/chemical synthesis , Organ Size , Phosphorothioate Oligonucleotides/administration & dosage , Phosphorothioate Oligonucleotides/chemical synthesis , Predictive Value of Tests , Quantitative Structure-Activity Relationship
3.
Nucleic Acids Res ; 38(20): 7100-11, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20615897

ABSTRACT

The potency and specificity of locked nucleic acid (LNA) antisense oligonucleotides was investigated as a function of length and affinity. The oligonucleotides were designed to target apolipoprotein B (apoB) and were investigated both in vitro and in vivo. The high affinity of LNA enabled the design of short antisense oligonucleotides (12- to 13-mers) that possessed high affinity and increased potency both in vitro and in vivo compared to longer oligonucleotides. The short LNA oligonucleotides were more target specific, and they exhibited the same biodistribution and tissue half-life as longer oligonucleotides. Pharmacology studies in both mice and non-human primates were conducted with a 13-mer LNA oligonucleotide against apoB, and the data showed that repeated dosing of the 13-mer at 1-2 mg/kg/week was sufficient to provide a significant and long lasting lowering of non-high-density lipoprotein (non-HDL) cholesterol without increasing serum liver toxicity markers. The data presented here show that oligonucleotide length as a parameter needs to be considered in the design of antisense oligonucleotide and that potent short oligonucleotides with sufficient target affinity can be generated using the LNA chemistry. Conclusively, we present a 13-mer LNA oligonucleotide with therapeutic potential that produce beneficial cholesterol lowering effect in non-human primates.


Subject(s)
Apolipoproteins B/metabolism , Cholesterol/blood , Oligonucleotides, Antisense/pharmacology , Oligonucleotides/pharmacology , Animals , Apolipoproteins B/genetics , Autoradiography , Base Pair Mismatch , Cell Line, Tumor , Female , Humans , Macaca fascicularis , Mice , Mice, Inbred C57BL , Oligonucleotides/chemistry , Oligonucleotides/pharmacokinetics , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacokinetics , RNA, Messenger/metabolism
4.
Neurobiol Dis ; 38(1): 36-46, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20053377

ABSTRACT

Inflammation induced by hypoxia-ischemia (HI) contributes to the development of injury in the newborn brain. In this study, we investigated the role of galectin-3, a novel inflammatory mediator, in the inflammatory response and development of brain injury in a mouse model for neonatal HI. Galectin-3 gene and protein expression was increased after injury and galectin-3 was located in activated microglia/macrophages. Galectin-3-deficient mice (gal3-/-) were protected from injury particularly in hippocampus and striatum. Microglia accumulation was increased in the gal3-/- mice but accompanied by decreased levels of total matrix metalloproteinase (MMP)-9 and nitrotyrosine. The protection and increase in microglial infiltration was more pronounced in male gal3-/- mice. Trophic factors and apoptotic markers did not significantly differ between groups. In conclusion, galectin-3 contributes to neonatal HI injury particularly in male mice. Our results indicate that galectin-3 exerts its effect by modulating the inflammatory response.


Subject(s)
Brain/metabolism , Encephalitis/metabolism , Galectin 3/metabolism , Hypoxia-Ischemia, Brain/metabolism , Inflammation Mediators/metabolism , Animals , Animals, Newborn , Brain/growth & development , Brain/physiopathology , Disease Models, Animal , Encephalitis/genetics , Encephalitis/physiopathology , Female , Galectin 3/genetics , Gliosis/genetics , Gliosis/metabolism , Gliosis/physiopathology , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/physiopathology , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Tyrosine/analogs & derivatives , Tyrosine/metabolism
5.
Nucleic Acids Res ; 38(1): e3, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19854938

ABSTRACT

For the past 15-20 years, the intracellular delivery and silencing activity of oligodeoxynucleotides have been essentially completely dependent on the use of a delivery technology (e.g. lipofection). We have developed a method (called 'gymnosis') that does not require the use of any transfection reagent or any additives to serum whatsoever, but rather takes advantage of the normal growth properties of cells in tissue culture in order to promote productive oligonucleotide uptake. This robust method permits the sequence-specific silencing of multiple targets in a large number of cell types in tissue culture, both at the protein and mRNA level, at concentrations in the low micromolar range. Optimum results were obtained with locked nucleic acid (LNA) phosphorothioate gap-mers. By appropriate manipulation of oligonucleotide dosing, this silencing can be continuously maintained with little or no toxicity for >240 days. High levels of oligonucleotide in the cell nucleus are not a requirement for gene silencing, contrary to long accepted dogma. In addition, gymnotic delivery can efficiently deliver oligonucleotides to suspension cells that are known to be very difficult to transfect. Finally, the pattern of gene silencing of in vitro gymnotically delivered oligonucleotides correlates particularly well with in vivo silencing. The establishment of this link is of particular significance to those in the academic research and drug discovery and development communities.


Subject(s)
Gene Silencing , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides/administration & dosage , Animals , Cell Line, Tumor , Humans , Indicators and Reagents , Mice , Oligonucleotides/analysis , Oligonucleotides, Antisense/analysis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Transfection
6.
Dev Neurosci ; 31(5): 420-6, 2009.
Article in English | MEDLINE | ID: mdl-19672071

ABSTRACT

Hypoxic-ischemic brain injury is often delayed and involves both apoptotic and immunoregulatory mechanisms. In this study, we used a neonatal model of hypoxia-ischemia to examine the effect of the mixed lineage kinase (MLK) inhibitor CEP-1347 on brain damage, apoptosis and inflammation. The tissue volume loss was reduced by 28% (p = 0.019) in CEP-1347-treated versus vehicle-treated rats and CEP-1347 significantly attenuated microgliosis at 7 days (p = 0.038). CEP-1347 decreased TUNEL-positive staining as well as cleaved caspase 3 immunoreactivity. CEP-1347 did not affect the expression of pro-inflammatory cytokines IL-1 beta, IL-6 and MCP-1, nor did it affect the expression of OX-42 (CR3) and OX-18 (MHC I) 24 h after the insult. In conclusion, the MLK inhibitor CEP-1347 has protective effects in a neonatal rat model of hypoxia-ischemia, which is mainly related to reduced apoptosis.


Subject(s)
Apoptosis/drug effects , Brain/pathology , Carbazoles/pharmacology , Hypoxia-Ischemia, Brain/drug therapy , Microglia/metabolism , Animals , Animals, Newborn , Blotting, Western , Brain/drug effects , Brain/metabolism , Caspase 3/metabolism , Cell Count , Enzyme-Linked Immunosorbent Assay , Hypoxia-Ischemia, Brain/metabolism , Image Processing, Computer-Assisted , Immunohistochemistry , In Situ Nick-End Labeling , Inflammation/metabolism , Microglia/drug effects , Neurons/drug effects , Neurons/metabolism , Organ Size/drug effects , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Statistics, Nonparametric
7.
ALTEX ; 26(2): 83-94, 2009.
Article in English | MEDLINE | ID: mdl-19565166

ABSTRACT

The role of microglia in neurodegeneration, toxicology and immunity is an expanding area of biomedical research requiring large numbers of animals. Use of a microglia-like cell line would accelerate many research programmes and reduce the necessity of continuous cell preparations and animal experimentation, provided that the cell line reproduces the in vivo situation or primary microglia (PM) with high fidelity. The immortalised murine microglial cell line BV-2 has been used frequently as a substitute for PM, but recently doubts were raised as to their suitability. Here, we re-evaluated strengths and potential short-comings of BV-2 cells. Their response to lipopolysaccharide was compared with the response of microglia in vitro and in vivo. Transcriptome (480 genes) and proteome analyses after stimulation with lipopolysaccharide indicated a reaction pattern of BV-2 with many similarities to that of PM, although the average upregulation of genes was less pronounced. The cells showed a normal regulation of NO production and a functional response to IFN-gamma, important parameters for appropriate interaction with T cells and neurons. BV-2 were also able to stimulate other glial cells. They triggered the translocation of NF-kappaB, and a subsequent production of IL-6 in astrocytes. Thus, BV-2 cells appear to be a valid substitute for PM in many experimental settings, incuding complex cell-cell interaction studies.


Subject(s)
Animal Testing Alternatives/methods , Brain/pathology , Inflammation/pathology , Microglia/cytology , Microglia/physiology , Animals , Astrocytes/physiology , Cell Line , Culture Media, Conditioned , Cytokines/metabolism , Gene Expression Regulation/physiology , Lipopolysaccharides/toxicity , Mice , NF-kappa B/metabolism , Nitrites/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Nucleic Acids Res ; 37(17): 5784-92, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19596814

ABSTRACT

microRNA-155 (miR-155) has been implicated as a central regulator of the immune system, but its function during acute inflammatory responses is still poorly understood. Here we show that exposure of cultured macrophages and mice to lipopolysaccharide (LPS) leads to up-regulation of miR-155 and that the transcription factor c/ebp Beta is a direct target of miR-155. Interestingly, expression profiling of LPS-stimulated macrophages combined with overexpression and silencing of miR-155 in murine macrophages and human monocytic cells uncovered marked changes in the expression of granulocyte colony-stimulating factor (G-CSF), a central regulator of granulopoiesis during inflammatory responses. Consistent with these data, we show that silencing of miR-155 in LPS-treated mice by systemically administered LNA-antimiR results in derepression of the c/ebp Beta isoforms and down-regulation of G-CSF expression in mouse splenocytes. Finally, we report for the first time on miR-155 silencing in vivo in a mouse inflammation model, which underscores the potential of miR-155 antagonists in the development of novel therapeutics for treatment of chronic inflammatory diseases.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/genetics , Gene Expression Regulation , Granulocyte Colony-Stimulating Factor/genetics , Inflammation/genetics , MicroRNAs/physiology , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line , Down-Regulation , Female , Gene Silencing , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/biosynthesis , Protein Biosynthesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Spleen/immunology
9.
Neurobiol Dis ; 31(1): 133-44, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18571099

ABSTRACT

BACKGROUND: Inflammation and reactive oxygen species (ROS) are important in the development of perinatal brain injury. The ROS-generating enzyme NADPH oxidase (Nox2) is present in inflammatory cells and contributes to brain injury in adult animal models. HYPOTHESIS: NADPH oxidase contributes to ROS formation and development of injury in the immature brain and inhibition of NADPH oxidase attenuates perinatal brain injury. METHODS: We used animal models of term hypoxia-ischemia (HI) (P9 mice) as well as ibotenate-induced excitotoxic injury (P5 mice) mimicking features of periventricular leukomalacia in preterm infants. In vitro microglia cell cultures were used to investigate NADPH oxidase-dependent ROS formation. In vivo we determined the impact 1) of HI on NADPH oxidase gene expression 2) of genetic (gp91-phox/Nox2 knock-out) and 3) of pharmacological NADPH oxidase inhibition on HI-induced injury and NMDA receptor-mediated excitotoxic injury, respectively. Endpoints were ROS formation, oxidative stress, apoptosis, inflammation and extent of injury. RESULTS: Hypoxia-ischemia increased NADPH oxidase subunits mRNA expression in total brain tissue in vivo. In vitro ibotenate increased NADPH oxidase-dependent formation of reactive oxygen species in microglia. In vivo the inhibition of NADPH oxidase did not reduce the extent of brain injury in any of the animal models. In contrast, the injury was increased by inhibition of NADPH oxidase and genetic inhibition was associated with an increased level of galectin-3 and IL-1beta. CONCLUSION: NADPH oxidase is upregulated after hypoxia-ischemia and activated microglia cells are a possible source of Nox2-derived ROS. In contrast to findings in adult brain, NADPH oxidase does not significantly contribute to the pathogenesis of perinatal brain injury. Results obtained in adult animals cannot be transferred to newborns and inhibition of NADPH oxidase should not be used in attempts to attenuate injury.


Subject(s)
Brain Injuries/enzymology , Hypoxia-Ischemia, Brain/enzymology , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Animals , Animals, Newborn , Apoptosis/physiology , Brain Injuries/etiology , Brain Injuries/pathology , Excitatory Amino Acid Agonists/toxicity , Female , Gene Expression , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/pathology , Ibotenic Acid/toxicity , Immunohistochemistry , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Knockout , Microglia/metabolism , NADPH Oxidases/genetics , Oxidative Stress/physiology , RNA, Messenger/analysis
10.
Nature ; 452(7189): 896-9, 2008 Apr 17.
Article in English | MEDLINE | ID: mdl-18368051

ABSTRACT

microRNAs (miRNAs) are small regulatory RNAs that are important in development and disease and therefore represent a potential new class of targets for therapeutic intervention. Despite recent progress in silencing of miRNAs in rodents, the development of effective and safe approaches for sequence-specific antagonism of miRNAs in vivo remains a significant scientific and therapeutic challenge. Moreover, there are no reports of miRNA antagonism in primates. Here we show that the simple systemic delivery of a unconjugated, PBS-formulated locked-nucleic-acid-modified oligonucleotide (LNA-antimiR) effectively antagonizes the liver-expressed miR-122 in non-human primates. Acute administration by intravenous injections of 3 or 10 mg kg(-1) LNA-antimiR to African green monkeys resulted in uptake of the LNA-antimiR in the cytoplasm of primate hepatocytes and formation of stable heteroduplexes between the LNA-antimiR and miR-122. This was accompanied by depletion of mature miR-122 and dose-dependent lowering of plasma cholesterol. Efficient silencing of miR-122 was achieved in primates by three doses of 10 mg kg(-1) LNA-antimiR, leading to a long-lasting and reversible decrease in total plasma cholesterol without any evidence for LNA-associated toxicities or histopathological changes in the study animals. Our findings demonstrate the utility of systemically administered LNA-antimiRs in exploring miRNA function in rodents and primates, and support the potential of these compounds as a new class of therapeutics for disease-associated miRNAs.


Subject(s)
Chlorocebus aethiops/genetics , Gene Silencing , MicroRNAs/genetics , Oligonucleotides/genetics , Animals , Female , Mice , Mice, Inbred C57BL , Oligonucleotides/administration & dosage , Oligonucleotides/adverse effects
11.
Nucleic Acids Res ; 36(4): 1153-62, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18158304

ABSTRACT

MicroRNA-122 (miR-122) is an abundant liver-specific miRNA, implicated in fatty acid and cholesterol metabolism as well as hepatitis C viral replication. Here, we report that a systemically administered 16-nt, unconjugated LNA (locked nucleic acid)-antimiR oligonucleotide complementary to the 5' end of miR-122 leads to specific, dose-dependent silencing of miR-122 and shows no hepatotoxicity in mice. Antagonism of miR-122 is due to formation of stable heteroduplexes between the LNA-antimiR and miR-122 as detected by northern analysis. Fluorescence in situ hybridization demonstrated uptake of the LNA-antimiR in mouse liver cells, which was accompanied by markedly reduced hybridization signals for mature miR-122 in treated mice. Functional antagonism of miR-122 was inferred from a low cholesterol phenotype and de-repression within 24 h of 199 liver mRNAs showing significant enrichment for miR-122 seed matches in their 3' UTRs. Expression profiling extended to 3 weeks after the last LNA-antimiR dose revealed that most of the changes in liver gene expression were normalized to saline control levels coinciding with normalized miR-122 and plasma cholesterol levels. Combined, these data suggest that miRNA antagonists comprised of LNA are valuable tools for identifying miRNA targets in vivo and for studying the biological role of miRNAs and miRNA-associated gene-regulatory networks in a physiological context.


Subject(s)
Gene Silencing , Liver/metabolism , MicroRNAs/antagonists & inhibitors , Oligonucleotides/pharmacology , RNA, Messenger/metabolism , Animals , Base Sequence , Female , Gene Expression Profiling , HeLa Cells , Humans , Liver/drug effects , Mice , MicroRNAs/chemistry , MicroRNAs/metabolism , Oligonucleotides/administration & dosage , Oligonucleotides/toxicity , Sequence Alignment , Up-Regulation
12.
J Neuroimmunol ; 180(1-2): 71-87, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16996144

ABSTRACT

Overall, the inflammatory potential of lipopolysaccharide (LPS) in vitro and in vivo was investigated using different omics technologies. We investigated the hippocampal response to intracerebroventricular (i.c.v) LPS in vivo, at both the transcriptional and protein level. Here, a time course analysis of interleukin-6 (IL-6) and monocyte chemotactic protein-1 (MCP-1) showed a sharp peak at 4 h and a return to baseline at 16 h. The expression of inflammatory mediators was not temporally correlated with expression of the microglia marker F4/80, which did not peak until 2 days after LPS injection. Of 480 inflammation-related genes present on a microarray, 29 transcripts were robustly up-regulated and 90% of them were also detected in LPS stimulated primary microglia (PM) cultures. Further in vitro to in vivo comparison showed that the counter regulation response observed in vivo was less evident in vitro, as transcript levels in PM decreased relatively little over 16 h. This apparent deficiency of homeostatic control of the innate immune response in cultures may also explain why a group of genes comprising tnf receptor associated factor-1, endothelin-1 and schlafen-1 were regulated strongly in vitro, but not in vivo. When the overall LPS-induced transcriptional response of PM was examined on a large Affymetrix chip, chemokines and cytokines constituted the most strongly regulated and largest groups. Interesting new microglia markers included interferon-induced protein with tetratricopeptide repeat (ifit), immune responsive gene-1 (irg-1) and thymidylate kinase family LPS-inducible member (tyki). The regulation of the former two was confirmed on the protein level in a proteomics study. Furthermore, conspicuous regulation of several gene clusters was identified, for instance that of genes pertaining to the extra-cellular matrix and enzymatic regulation thereof. Although most inflammatory genes induced in vitro were transferable to our in vivo model, the observed discrepancy for some genes potentially represents regulatory factors present in the central nervous system (CNS) but not in vitro.


Subject(s)
Encephalitis/physiopathology , Gene Expression/drug effects , Gliosis/physiopathology , Lipopolysaccharides/pharmacology , Microglia/drug effects , Transcriptional Activation/drug effects , Animals , Animals, Newborn , Antigens, Differentiation/immunology , Antigens, Differentiation/metabolism , Cells, Cultured , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Disease Models, Animal , Encephalitis/chemically induced , Encephalitis/immunology , Gene Expression/immunology , Gene Expression Profiling , Gliosis/chemically induced , Gliosis/immunology , Hippocampus/drug effects , Hippocampus/immunology , Hippocampus/physiopathology , Inflammation Mediators/pharmacology , Injections, Intraventricular , Interleukin-6/immunology , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/immunology , Oligonucleotide Array Sequence Analysis , Proteomics , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Transcriptional Activation/immunology , Up-Regulation/drug effects , Up-Regulation/immunology
13.
Am J Pathol ; 169(3): 967-76, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16936270

ABSTRACT

Preterm birth is a major contributor of adverse perinatal outcome. Clinical data suggest that an inflammatory response is important in the process leading to preterm labor. By using a recently introduced mouse model of localized intrauterine lipopolysaccharide-induced inflammation, the effect of interleukin (IL)-18 gene disruption and/or IL-18 neutralization as well as combined IL-1alpha/beta gene disruption on inflammation-induced fetal loss was investigated. The frequency of preterm fetal loss was significantly higher in IL-18 knockout mice (58.9%) and in mice administered IL-18-binding protein (59.7%) compared to wild-type controls (34.7%). The rate of fetal loss was not affected by IL-1alpha/beta gene deficiency (38.7%). Decreased IL-18 protein expression combined with elevated IL-12 protein expression in uterine tissue of IL-18 knockout mice and IL-18-binding protein-treated animals was noticed. These data demonstrate that preterm pregnancy loss in response to intrauterine inflammation was enhanced by disruption of the IL-18 gene and/or IL-18 neutralization, events that may relate to exaggerated Th1 responses because of an increased IL-12/IL-18 ratio.


Subject(s)
Fetal Death/genetics , Interleukin-18/genetics , Interleukin-1/deficiency , Animals , Female , Fetal Death/chemically induced , Fetal Death/immunology , Fetal Death/pathology , Glycoproteins/immunology , Glycoproteins/pharmacology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Intercellular Signaling Peptides and Proteins , Interleukin-1/immunology , Interleukin-12/immunology , Interleukin-18/immunology , Lipopolysaccharides/pharmacology , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , Pregnancy , Th1 Cells/immunology , Th1 Cells/pathology , Uterus/immunology , Uterus/pathology
14.
Pediatr Res ; 60(2): 161-8, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16864697

ABSTRACT

To improve the understanding of the molecular mechanisms whereby lipopolysaccharide (LPS) affects the immature brain, global gene expression following LPS exposure was investigated in neonatal rats. Brains (n = 5/time point) were sampled 2, 6, and 72 h after LPS and compared with age-matched controls. The mRNA from each brain was analyzed separately on Affymextrix GeneChip Rat Expression Set 230. The number of genes regulated after LPS were 847 at 2 h, 1564 at 6 h, and 1546 genes at 72 h. Gene ontology analysis demonstrated that, at both 2 and 6 h after LPS, genes associated with protein metabolism, response to external stimuli and stress (immune and inflammatory response, chemotaxis) and cell death were overrepresented. At 72 h, the most strongly regulated genes belonged to secretion of neurotransmitters, transport, synaptic transmission, cell migration, and neurogenesis. Several pathways associated with cell death/survival were identified (caspase-tumor necrosis factor alpha [TNF-alpha]-, p53-, and Akt/phosphatidylinositol-3-kinase (PI3 K)-dependent mechanisms). Caspase-3 activity increased and phosphorylation of Akt decreased 8 h after peripheral LPS exposure. These results show a complex cerebral response to peripheral LPS exposure. In addition to the inflammatory response, a significant number of cell death-associated genes were identified, which may contribute to increased vulnerability of the immature brain to hypoxia-ischemia (HI) following LPS exposure.


Subject(s)
Apoptosis/genetics , Brain/metabolism , Gene Expression/drug effects , Inflammation/genetics , Lipopolysaccharides/toxicity , Animals , Animals, Newborn , Brain/drug effects , Gene Expression Profiling , Rats , Rats, Wistar
15.
Dev Neurosci ; 27(2-4): 143-8, 2005.
Article in English | MEDLINE | ID: mdl-16046848

ABSTRACT

Interleukin (IL)-1 and IL-18 belong to the IL-1 family. IL-18 deficiency has been shown to confer moderate protection after hypoxia-ischemia (HI) in the immature brain, while the contribution of the two isoforms of IL-1 (IL-1alpha and IL-1beta) in neonatal HI brain injury has not been investigated previously. The aim of this study was to examine the contribution of the different members of the IL-1 family to neonatal HI damage. Unilateral HI was induced at postnatal day 9 in IL-1beta, IL-1beta18, and IL-1alphabeta knockout and wild-type mice and brain injury was evaluated 1 week later. IL-1beta18-deficient mice showed 17% reduction in brain injury, while no significant reduction in injury was detected between any of the other groups. These results indicate that IL-18, but not IL-1beta, or the combination of IL-1alpha and IL-1beta, is a contributor to HI injury in the immature brain.


Subject(s)
Hypoxia-Ischemia, Brain/metabolism , Interleukin-18/metabolism , Interleukin-1/metabolism , Animals , Animals, Newborn , Brain/blood supply , Brain/metabolism , Brain/pathology , DNA Primers , Disease Susceptibility , Hypoxia-Ischemia, Brain/pathology , Mice , Mice, Knockout , Polymerase Chain Reaction
16.
Eur J Neurosci ; 21(6): 1489-502, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15845077

ABSTRACT

Insulin-like growth factor I (IGF-I) is a neurotrophic factor that promotes neuronal growth, differentiation and survival. Neuroprotective effects of IGF-I have previously been shown in adult and juvenile rat models of brain injury. We wanted to investigate the neuroprotective effect of IGF-I after hypoxia-ischemia (HI) in 7-day-old neonatal rats and the mechanisms of IGF-I actions in vivo. We also wanted to study effects of HI and/or IGF-I on the serine/threonine kinases Akt and glycogen synthase kinase 3beta (GSK3beta) in the phophatidylinositol-3 kinase (PI3K) pathway. Immediately after HI, phosphorylated Akt (pAkt) and phosphorylated GSK3beta (pGSK3beta) immunoreactivity was lost in the ipsilateral and reduced in the contralateral hemisphere. After 45 min, pAkt levels were restored to control values, whereas pGSK3beta remained low 4 h after HI. Administration of IGF-I (50 microg i.c.v.) after HI resulted in a 40% reduction in brain damage (loss of microtubule-associated protein) compared with vehicle-treated animals. IGF-I treatment without HI was shown to increase pAkt whereas pGSK3beta decreased in the cytosol, but increased in the nuclear fraction. IGF-I treatment after HI increased pAkt in the cytosol and pGSK3beta in both the cytosol and the nuclear fraction in the ipsilateral hemisphere compared with vehicle-treated rats, concomitant with a reduced caspase-3- and caspase-9-like activity. In conclusion, IGF-I induces activation of Akt during recovery after HI which, in combination with inactivation of GSK3beta, may explain the attenuated activation of caspases and reduction of injury in the immature brain.


Subject(s)
Glycogen Synthase Kinase 3/physiology , Hypoxia-Ischemia, Brain/prevention & control , Insulin-Like Growth Factor I/physiology , Insulin-Like Growth Factor I/therapeutic use , Neuroprotective Agents/therapeutic use , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Animals , Animals, Newborn , Brain/drug effects , Brain/growth & development , Brain/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/metabolism , Neuroprotective Agents/pharmacology , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Rats , Rats, Wistar
17.
Neurosci Lett ; 373(1): 16-20, 2005 Jan 03.
Article in English | MEDLINE | ID: mdl-15555769

ABSTRACT

Inflammation is likely to be important in the pathophysiology of white matter damage in the immature brain. In order to investigate the involvement of interleukin (IL)-18, we subjected 9-day-old IL-18-deficient and wild-type (WT) mice to hypoxia-ischemia (HI) (unilateral carotid ligation and exposure to 10% oxygen) and white matter injury was evaluated after 3 days by immunostaining for myelin basic protein (MBP) and neurofilament (NF). The immunoreactivity of MBP was significantly higher by 92, 49 and 21%, respectively, in subcortical white matter, striatum and thalamus in IL-18-deficient mice versus WT mice following HI. Similarly, there was a more pronounced immunoreactivity of NF by 78% in the subcortical white matter in IL-18 KO versus WT mice. IL-18 was expressed by astrocytes and microglia, whereas the IL-18 receptor was mainly found in astrocytes localized in and around the subventricular white matter. Taken together, these results indicate that release of IL-18 may play an important role in the development of white matter injury in the neonatal brain.


Subject(s)
Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Interleukin-18/metabolism , Animals , Animals, Newborn , Astrocytes/metabolism , Female , Immunohistochemistry , Interleukin-18/deficiency , Male , Mice , Mice, Knockout , Microglia/metabolism , Myelin Basic Protein/metabolism
18.
J Cereb Blood Flow Metab ; 24(12): 1317-32, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15625407

ABSTRACT

Ischemia induces a complex response of differentially expressed genes in the brain. In order to understand the specific mechanisms of injury in the developing brain, it is important to obtain information on global changes in the transcriptome after neonatal hypoxia-ischemia. In this study, oligonucleotide arrays were used to investigate genomic changes at 2, 8, 24, and 72 hours after neonatal hypoxia-ischemia, which was induced in 9-day-old mice by left carotid artery ligation followed by hypoxia (10% O2). In total, 343 genes were differentially expressed in cortex, hippocampus, thalamus, and striatum 2 to 72 hours after hypoxia-ischemia, when comparing ipsilateral with contralateral hemispheres and with controls, using the significance analysis for microarrays. A total of 283 genes were upregulated and 60 were downregulated, and 94% of the genes had not previously been shown after neonatal hypoxia-ischemia. Genes related to transcription factors and metabolism had mostly upregulated transcripts, whereas most downregulated genes belonged to the categories of ion and vesicular transport and signal transduction. Genes involved in transcription, stress, and apoptosis were induced early after the insult, and many new genes that may play important roles in the pathophysiology of neonatal hypoxia-ischemia were identified.


Subject(s)
Aging/genetics , Gene Expression Profiling , Hypoxia-Ischemia, Brain/genetics , Animals , Animals, Newborn , Apoptosis/genetics , Cytoskeleton/genetics , Gene Expression Regulation , Growth Substances/genetics , Hormones/genetics , Mice , Mice, Inbred C57BL , Neurotransmitter Agents/genetics , Oligonucleotide Array Sequence Analysis , Protein Transport , RNA, Messenger/analysis , RNA, Messenger/genetics , Signal Transduction , Stress, Physiological/genetics , Synapses/genetics , Transcription, Genetic/genetics
19.
J Cereb Blood Flow Metab ; 24(12): 1333-51, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15625408

ABSTRACT

Brain ischemia triggers an inflammatory reaction that progresses for days to weeks and seems to have a role in secondary progression of injury. Inflammation induces a complex pattern of signaling molecules with partly contradictory actions, and the responses may be different in the immature and adult brain. The authors characterized the global inflammatory gene expression in the developing brain as a first step toward understanding the protective and deleterious effects of inflammation after hypoxia-ischemia. Oligonucleotide arrays were used to investigate inflammatory genes in cortex, hippocampus, thalamus, and striatum at 2, 8, 24, and 72 hours after hypoxia-ischemia, which was induced in 9-day-old mice by left carotid artery ligation followed by hypoxia. After hypoxia-ischemia, 148 inflammatory genes were differentially expressed. More than 97% of the genes were upregulated and 93% had not previously been reported after hypoxia-ischemia in the immature brain. The results indicate that microglia/macrophages, T- and B-cells, NK-cells, mast cells, dendritic cells, and polymorphonuclear leukocytes may participate in the response to hypoxia-ischemia. In addition, novel cytokines/chemokines, complement-related, interferon-regulated, components of the TIR/nuclear factor-kappaB pathway, and a number of immunomodulatory genes were induced. Several of these genes may of pathophysiologic significance after neonatal hypoxia-ischemia.


Subject(s)
Aging/genetics , Gene Expression Profiling , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/pathology , Inflammation/genetics , Animals , Cell Adhesion/genetics , Chemokines/genetics , Gene Expression Regulation , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class II/genetics , Immunohistochemistry , Interferons/genetics , Leukocytes/metabolism , Macrophages/metabolism , Mice
20.
J Neurosci ; 22(14): 5910-9, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-12122053

ABSTRACT

Inflammation is a critical factor for development of hypoxic-ischemic (HI) brain injury. Interleukin-18 (IL-18) is a proinflammatory cytokine expressed in microglia and processed by caspase-1. Our aim was to characterize the expression of IL-18 and its receptor in relation to caspase-1 and IL-1beta after HI and to evaluate to what extent IL-18 contributes to HI brain injury. Seven-day-old rats were subjected to HI, and brain tissue was sampled at different time points (3 hr to 14 d) after insult. The mRNA for IL-18 and caspase-1 were analyzed with reverse transcriptase PCR, protein was analyzed by Western blot (IL-18, caspase-1) or ELISA (IL-1beta), and the regional distribution was assessed by immunohistochemistry. HI was also induced in C57BL/6 mice, and brain injury in IL-18-deficient animals was compared with that in wild-type animals. The expression of mRNA/protein for caspase-1 and IL-18 in brain homogenates increased progressively at 12 hr to 14 d after HI, whereas IL-1beta peaked at 8 hr. A widespread expression of caspase-1 and IL-18 protein in microglia was found in the HI hemisphere. The IL-18 receptor was expressed on neurons of the cerebral cortex and thalamus. IL-1beta was primarily found in microglia in the habenular nucleus of the thalamus. The infarct volume was reduced by 21% (p = 0.01), and the neuropathology score was significantly decreased in the cerebral cortex (-35%), hippocampus (-22%), striatum (-18%), and thalamus (-17%) in mice with IL-18 deficiency compared with wild-type mice. In conclusion, we found that IL-18 expression in microglia was markedly increased after HI and that IL-18 appears to be important for the development of HI brain injury.


Subject(s)
Hypoxia-Ischemia, Brain/metabolism , Interleukin-18/metabolism , Animals , Animals, Newborn , Blotting, Western , Brain/blood supply , Brain/metabolism , Brain/pathology , Brain Chemistry , Caspase 1/genetics , Caspase 1/metabolism , Disease Models, Animal , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Hypoxia-Ischemia, Brain/pathology , Immunohistochemistry , Interleukin-1/metabolism , Interleukin-18/deficiency , Interleukin-18/genetics , Interleukin-18 Receptor alpha Subunit , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Interleukin/biosynthesis , Receptors, Interleukin-18 , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...