Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Neurobiol Dis ; 148: 105200, 2021 01.
Article in English | MEDLINE | ID: mdl-33248237

ABSTRACT

Hypoxia-inducible factor-1α (HIF1α) is a major regulator of cellular adaptation to hypoxia and oxidative stress, and recent advances of prolyl-4-hydroxylase (P4H) inhibitors have produced powerful tools to stabilize HIF1α for clinical applications. However, whether HIF1α provokes or resists neonatal hypoxic-ischemic (HI) brain injury has not been established in previous studies. We hypothesize that systemic and brain-targeted HIF1α stabilization may have divergent effects. To test this notion, herein we compared the effects of GSK360A, a potent P4H inhibitor, in in-vitro oxygen-glucose deprivation (OGD) and in in-vivo neonatal HI via intracerebroventricular (ICV), intraperitoneal (IP), and intranasal (IN) drug-application routes. We found that GSK360A increased the erythropoietin (EPO), heme oxygenase-1 (HO1) and glucose transporter 1 (Glut1) transcripts, all HIF1α target-genes, and promoted the survival of neurons and oligodendrocytes after OGD. Neonatal HI insult stabilized HIF1α in the ipsilateral hemisphere for up to 24 h, and either ICV or IN delivery of GSK360A after HI increased the HIF1α target-gene transcripts and decreased brain damage. In contrast, IP-injection of GSK360A failed to reduce HI brain damage, but elevated the risk of mortality at high doses, which may relate to an increase of the kidney and plasma EPO, leukocytosis, and abundant vascular endothelial growth factor (VEGF) mRNAs in the brain. These results suggest that brain-targeted HIF1α-stabilization is a potential treatment of neonatal HI brain injury, while systemic P4H-inhibition may provoke unwanted adverse effects.


Subject(s)
Enzyme Inhibitors/pharmacology , Glycine/analogs & derivatives , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Ischemia, Brain/metabolism , Neurons/drug effects , Oligodendroglia/drug effects , Quinolones/pharmacology , Administration, Intranasal , Animals , Animals, Newborn , Cell Survival/drug effects , Erythropoietin/genetics , Glucose Transporter Type 1/drug effects , Glucose Transporter Type 1/genetics , Glycine/pharmacology , Heme Oxygenase (Decyclizing)/drug effects , Heme Oxygenase (Decyclizing)/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Injections, Intraperitoneal , Injections, Intraventricular , Neurons/metabolism , Oligodendroglia/metabolism , Rats
2.
Cell Cycle ; 14(23): 3698-712, 2015.
Article in English | MEDLINE | ID: mdl-26505089

ABSTRACT

Traumatic spinal cord injury (SCI) induces cell cycle activation (CCA) that contributes to secondary injury and related functional impairments such as motor deficits and hyperpathia. E2F1 and E2F2 are members of the activator sub-family of E2F transcription factors that play an important role in proliferating cells and in cell cycle-related neuronal death, but no comprehensive study have been performed in SCI to determine the relative importance of these factors. Here we examined the temporal distribution and cell-type specificity of E2F1 and E2F2 expression following mouse SCI, as well as the effects of genetic deletion of E2F1-2 on neuronal cell death, neuroinflammation and associated neurological dysfunction. SCI significantly increased E2F1 and E2F2 expression in active caspase-3(+) neurons/oligodendrocytes as well as in activated microglia/astrocytes. Injury-induced up-regulation of cell cycle-related genes and protein was significantly reduced by intrathecal injection of high specificity E2F decoy oligodeoxynucleotides against the E2F-binding site or in E2F1-2 null mice. Combined E2F1+2 siRNA treatment show greater neuroprotection in vivo than E2F1 or E2F2 single siRNA treatment. Knockout of both E2F1 and E2F2 genes (E2Fdko) significantly reduced neuronal death, neuroinflammation, and tissue damage, as well as limiting motor dysfunction and hyperpathia after SCI. Both CCA reduction and functional improvement in E2Fdko mice were greater than those in E2F2ko model. These studies demonstrate that SCI-induced activation of E2F1-2 mediates CCA, contributing to gliopathy and neuronal/tissue loss associated with motor impairments and post-traumatic hyperesthesia. Thus, E2F1-2 provide a therapeutic target for decreasing secondary tissue damage and promoting recovery of function after SCI.


Subject(s)
E2F1 Transcription Factor/physiology , E2F2 Transcription Factor/physiology , Spinal Cord Injuries/pathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cell Death , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism , Gene Expression , Gene Knockout Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism
3.
CNS Neurosci Ther ; 21(4): 367-73, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25475942

ABSTRACT

Hypoxic-ischemic brain injury is an important cause of neurodevelopmental deficits in neonates. Intrauterine infection and the ensuing fetal inflammatory responses augment hypoxic-ischemic brain injury and attenuate the efficacy of therapeutic hypothermia. Here, we review evidences from preclinical studies suggesting that the induction of brain parenchymal tissue-type plasminogen activator (tPA) plays an important pathogenic role in these conditions. Moreover, administration of a stable-mutant form of plasminogen activator inhibitor-1 called CPAI confers potent protection against hypoxic-ischemic injury with and without inflammation via different mechanisms. Besides intracerebroventricular injection, CPAI can also be administered into the brain using a noninvasive intranasal delivery strategy, adding to its applicability in clinical use. In sum, the therapeutic potential of CPAI in neonatal care merits further investigation with large-animal models of hypoxia-ischemia and cerebral palsy.


Subject(s)
Encephalitis/drug therapy , Encephalitis/physiopathology , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/physiopathology , Neuroprotective Agents/administration & dosage , Plasminogen Inactivators/administration & dosage , Animals , Brain/drug effects , Brain/physiopathology , Humans , Tissue Plasminogen Activator/antagonists & inhibitors
4.
J Neurosci ; 34(49): 16467-81, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25471584

ABSTRACT

Intrauterine infection (chorioamnionitis) aggravates neonatal hypoxic-ischemic (HI) brain injury, but the mechanisms linking systemic inflammation to the CNS damage remain uncertain. Here we report evidence for brain influx of T-helper 17 (TH17)-like lymphocytes to coordinate neuroinflammatory responses in lipopolysaccharide (LPS)-sensitized HI injury in neonates. We found that both infants with histological chorioamnionitis and rat pups challenged by LPS/HI have elevated expression of the interleukin-23 (IL-23) receptor, a marker of early TH17 lymphocytes, in the peripheral blood mononuclear cells. Post-LPS/HI administration of FTY720 (fingolimod), a sphingosine-1-phosphate receptor agonist that blocks lymphocyte trafficking, mitigated the influx of leukocytes through the choroid plexus and acute induction of nuclear factor-κB signaling in the brain. Subsequently, the FTY720 treatment led to attenuated blood-brain barrier damage, fewer cluster of differentiation 4-positive, IL-17A-positive T-cells in the brain, less proinflammatory cytokine, and better preservation of growth and white matter functions. The FTY720 treatment also provided dose-dependent reduction of brain atrophy, rescuing >90% of LPS/HI-induced brain tissue loss. Interestingly, FTY720 neither opposed pure-HI brain injury nor directly inhibited microglia in both in vivo and in vitro models, highlighting its unique mechanism against inflammation-sensitized HI injury. Together, these results suggest that the dual hit of systemic inflammation and neonatal HI injury triggers early onset of the TH17/IL-17-mediated immunity, which causes severe brain destruction but responds remarkably to the therapeutic blockade of lymphocyte trafficking.


Subject(s)
Cell Movement/drug effects , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/prevention & control , Inflammation/prevention & control , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , Propylene Glycols/pharmacology , Sphingosine/analogs & derivatives , Animals , Animals, Newborn , Atrophy/drug therapy , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain/metabolism , Brain/pathology , Chorioamnionitis/drug therapy , Chorioamnionitis/metabolism , Cytokines/metabolism , Dose-Response Relationship, Drug , Female , Fingolimod Hydrochloride , Humans , Hypoxia-Ischemia, Brain/drug therapy , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Infant, Newborn , Lipopolysaccharides , Lymphocytes/cytology , NF-kappa B/metabolism , Pregnancy , Propylene Glycols/therapeutic use , Rats , Receptors, Interleukin/metabolism , Sphingosine/pharmacology , Sphingosine/therapeutic use , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , White Matter/drug effects
5.
PLoS One ; 9(6): e98807, 2014.
Article in English | MEDLINE | ID: mdl-24911517

ABSTRACT

Edaravone, a potent antioxidant, may improve thrombolytic therapy because it benefits ischemic stroke patients on its own and mitigates adverse effects of tissue plasminogen activator (tPA) in preclinical models. However, whether the combined tPA-edaravone therapy is more effective in reducing infarct size than singular treatment is uncertain. Here we investigated this issue using a transient hypoxia-ischemia (tHI)-induced thrombotic stroke model, in which adult C57BL/6 mice were subjected to reversible ligation of the unilateral common carotid artery plus inhalation of 7.5% oxygen for 30 min. While unilateral occlusion of the common carotid artery suppressed cerebral blood flow transiently, the addition of hypoxia triggered reperfusion deficits, endogenous thrombosis, and attenuated tPA activity, leading up to infarction. We compared the outcomes of vehicle-controls, edaravone treatment, tPA treatment at 0.5, 1, or 4 h post-tHI, and combined tPA-edaravone therapies with mortality rate and infarct size as the primary end-points. The best treatment was further compared with vehicle-controls in behavioral, biochemical, and diffusion tensor imaging (DTI) analyses. We found that application of tPA at 0.5 or 1 h--but not at 4 h post-tHI--significantly decreased infarct size and showed synergistic (p<0.05) or additive benefits with the adjuvant edaravone treatment, respectively. The acute tPA-edaravone treatment conferred >50% reduction of mortality, ∼ 80% decline in infarct size, and strong white-matter protection. It also improved vascular reperfusion and decreased oxidative stress, inflammatory cytokines, and matrix metalloproteinase activities. In conclusion, edaravone synergizes with acute tPA treatment in experimental thrombotic stroke, suggesting that clinical application of the combined tPA-edaravone therapy merits investigation.


Subject(s)
Antipyrine/analogs & derivatives , Intracranial Thrombosis/complications , Stroke/complications , Stroke/drug therapy , Tissue Plasminogen Activator/pharmacology , Animals , Antipyrine/pharmacology , Antipyrine/therapeutic use , Cell Hypoxia/drug effects , Disease Models, Animal , Drug Synergism , Edaravone , Humans , Hypoxia-Ischemia, Brain/complications , Male , Mice , Mice, Inbred C57BL , Stroke/etiology , Stroke/physiopathology , Tissue Plasminogen Activator/metabolism , Tissue Plasminogen Activator/therapeutic use , White Matter/drug effects , White Matter/injuries
6.
Sci Transl Med ; 5(193): 193ra90, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23843451

ABSTRACT

Intracranial hemorrhage in preterm neonates may result in neonatal mortality and functional disabilities, but its pathogenic mechanisms are poorly defined and better therapies are needed. We used a tetracycline-regulated transgenic system to test whether the induction of vascular endothelial growth factor (VEGF) in the germinal matrix leads to intracranial hemorrhage. This genetic strategy initially induced a dense network of loosely adjoined endothelial cells and pericytes near lateral ventricles, similar to the immature vascular rete in human fetal brains. Yet, this rich vascular network transformed into low-vasculature patches correlated with hemorrhage and caspase-3 activation near birth. Gene expression and biochemical analyses suggested that downstream mediators of VEGF in this network include transcriptional factors ETS1 and HIF2α (hypoxia-inducible factor 2α), components of the PDGFß (platelet-derived growth factor ß) and TGFß (transforming growth factor-ß) receptor signaling pathways, matrix metalloproteinase-9 (MMP-9), and cathepsins. Prenatal administration of glucocorticoids markedly reduced mortality and cerebral hemorrhage in mutant animals, as in human neonates. This protective effect was not due to blocking vasculogenesis, but was instead associated with inhibition of neurovascular proteases, notably MMP-9, cathepsin B, and caspase-3. Collectively, these results support a causative role of VEGF in perinatal cerebral hemorrhage and implicate its downstream proteases as potential therapeutic targets.


Subject(s)
Cerebral Hemorrhage/enzymology , Cerebral Hemorrhage/pathology , Peptide Hydrolases/biosynthesis , Prosencephalon/enzymology , Prosencephalon/pathology , Vascular Endothelial Growth Factor A/metabolism , Animals , Animals, Newborn , Betamethasone/pharmacology , Betamethasone/therapeutic use , Caspase 3/metabolism , Cathepsin B/metabolism , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/genetics , Disease Models, Animal , Embryo, Mammalian/drug effects , Embryo, Mammalian/pathology , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Gene Expression Profiling , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , Humans , Matrix Metalloproteinase 9/metabolism , Mice , Neovascularization, Pathologic/drug therapy , Phenotype , Prosencephalon/blood supply , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use , Tetracycline/pharmacology
7.
Stroke ; 44(9): 2623-2627, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23881953

ABSTRACT

BACKGROUND AND PURPOSE: Plasminogen activator inhibitor-I (PAI-1), a ≈50-kDa serine protease inhibitor, markedly reduces the extravascular toxicity of tissue-type plasminogen activator in experimental hypoxic-ischemic (HI) brain injury of newborns. However, the current treatment with PAI-1 requires intracerebroventricle injection to cross the blood-brain barrier, which is an invasive procedure of limited clinical potential. Thus, we tested whether intranasal administration of PAI-1 can bypass blood-brain barrier and mitigate neonatal HI brain injury. METHODS: Rat pups were subjected to HI, with or without lipopolysaccharide pre-exposure, followed by intranasal delivery of a stable-mutant form of PAI-1 (CPAI). RESULTS: Immunoblotting showed that CPAI sequentially entered the olfactory bulbs and cerebral cortex after intranasal delivery and reduced ≈75% of brain atrophy in HI or lipopolysaccharide-sensitized HI injury. Mechanistically, CPAI attenuated HI-induced plasminogen activators and lipopolysaccharide/HI-induced nuclear factor-κB signaling, neuroinflammation, and blood-brain barrier permeability. CONCLUSIONS: Intranasal delivery of CPAI is an effective treatment of experimental HI brain injury of newborns. Clinical application of this experimental therapy merits further investigation.


Subject(s)
Hypoxia-Ischemia, Brain/drug therapy , Plasminogen Activator Inhibitor 1/administration & dosage , Plasminogen Inactivators/administration & dosage , Plasminogen Inactivators/therapeutic use , Administration, Intranasal , Animals , Animals, Newborn , Atrophy , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Disease Models, Animal , Drug Administration Schedule , Female , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Male , Plasminogen Activator Inhibitor 1/therapeutic use , Rats , Rats, Wistar , Serine Proteinase Inhibitors/administration & dosage , Serine Proteinase Inhibitors/therapeutic use
8.
Genes Dev ; 27(11): 1272-87, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23723414

ABSTRACT

Neural stem cells (NSCs) reside in widespread regions along the lateral ventricle and generate diverse olfactory bulb (OB) interneuron subtypes in the adult mouse brain. Molecular mechanisms underlying their regional diversity, however, are not well understood. Here we show that the homeodomain transcription factor Gsx2 plays a crucial role in the region-specific control of adult NSCs in both persistent and injury-induced neurogenesis. In the intact brain, Gsx2 is expressed in a regionally restricted subset of NSCs and promotes the activation and lineage progression of stem cells, thereby controlling the production of selective OB neuron subtypes. Moreover, Gsx2 is ectopically induced in damaged brains outside its normal expression domains and is required for injury-induced neurogenesis in the subventricular zone (SVZ). These results demonstrate that mobilization of adult NSCs is controlled in a region-specific manner and that distinct mechanisms operate in continuous and injury-induced neurogenesis in the adult brain.


Subject(s)
Homeodomain Proteins/metabolism , Lateral Ventricles/cytology , Lateral Ventricles/injuries , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Adult Stem Cells/metabolism , Animals , Cell Lineage , Lateral Ventricles/metabolism , Mice , Neural Stem Cells/classification , Olfactory Bulb/cytology , Organ Specificity , Stem Cell Niche , Transcription Factors/metabolism
9.
Exp Neurol ; 247: 447-455, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23353638

ABSTRACT

Perinatal infection aggravates neonatal hypoxic-ischemic (HI) brain injury and may interfere with therapeutic hypothermia. While the NF-κB signaling pathway has been implicated in microglia activation in infection-sensitized HI, the current therapeutic strategies rely on systemic intervention, which could impair neonatal immunity and increase the risk of severe infection. To devise a brain-targeted anti-NF-κB strategy, we examined the effects of intranasal delivery of tat-NBD peptides in two animal models of neonatal infection-sensitized HI. Kinetic experiments showed that tat-NBD peptides entered the olfactory bulbs rapidly (10-30 min) and peaked in the cerebral cortex around 60 min after intranasal application in P7 rats. Further, intranasal delivery of 1.4 mg/kg tat-NBD, which is only 7% of the intravenous dose in past studies, markedly attenuated NF-κB signaling, microglia activation, and brain damage triggered by HI with 4 or 72 h pre-exposure to the bacterial endotoxin lipopolysaccharide (LPS). In contrast, intranasal delivery of mutant tat-NBD peptides or systemic application of minocycline failed to block LPS-sensitized HI injury. Yet, intranasal delivery of up to 5.6 mg/kg tat-NBD peptides immediately after pure-HI insult showed little protection, likely due to its rapid clearance from the brain and inability to inhibit parenchymal plasminogen activators. Together, these results suggest a novel therapy of infection-sensitized HI brain injury in newborns.


Subject(s)
Hypoxia-Ischemia, Brain/drug therapy , Intracellular Signaling Peptides and Proteins/administration & dosage , NF-kappa B/antagonists & inhibitors , Peptides/administration & dosage , Administration, Intranasal , Analysis of Variance , Animals , Animals, Newborn , Brain/drug effects , Brain/metabolism , Brain/pathology , CD11b Antigen/metabolism , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Electrophoretic Mobility Shift Assay , Female , Hypoxia-Ischemia, Brain/pathology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/pharmacology , Magnetic Resonance Spectroscopy , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Microfilament Proteins/metabolism , Mutation/physiology , Rats , Tetrazolium Salts
10.
Cereb Cortex ; 23(5): 1218-29, 2013 May.
Article in English | MEDLINE | ID: mdl-22556277

ABSTRACT

Intrauterine infection exacerbates neonatal hypoxic-ischemic (HI) brain injury and impairs the development of cerebral cortex. Here we used low-dose lipopolysaccharide (LPS) pre-exposure followed by unilateral cerebral HI insult in 7-day-old rats to study the pathogenic mechanisms. We found that LPS pre-exposure blocked the HI-induced proteolytic activity of tissue-type plasminogen activator (tPA), but significantly enhanced NF-κB signaling, microglia activation, and the production of pro-inflammatory cytokines in newborn brains. Remarkably, these pathogenic responses were all blocked by intracerebroventricular injection of a stable-mutant form of plasminogen activator protein-1 called CPAI. Similarly, LPS pre-exposure amplified, while CPAI therapy mitigated HI-induced blood-brain-barrier damage and the brain tissue loss with a therapeutic window at 4 h after the LPS/HI insult. The CPAI also blocks microglia activation following a brain injection of LPS, which requires the contribution by tPA, but not the urinary-type plasminogen activator (uPA), as shown by experiments in tPA-null and uPA-null mice. These results implicate the nonproteolytic tPA activity in LPS/HI-induced brain damage and microglia activation. Finally, the CPAI treatment protects near-normal motor and white matter development despite neonatal LPS/HI insult. Together, because CPAI blocks both proteolytic and nonproteolytic tPA neurotoxicity, it is a promising therapeutics of neonatal HI injury either with or without infection.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/prevention & control , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/prevention & control , Lipopolysaccharides , Plasminogen Activator Inhibitor 1/pharmacology , Tissue Plasminogen Activator/metabolism , Animals , Animals, Newborn , Encephalitis/chemically induced , Encephalitis/metabolism , Encephalitis/prevention & control , Hypoxia-Ischemia, Brain/chemically induced , Mice , Rats
11.
J Neurosci Methods ; 203(1): 122-9, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21982741

ABSTRACT

A simple method to quantify cerebral infarction has great value for mechanistic and therapeutic studies in experimental stroke research. Immersion staining of unfixed brain slices with 2,3,5-triphenyltetrazolium chloride (TTC) is a popular method to determine cerebral infarction in preclinical studies. However, it is often difficult to apply immersion TTC-labeling to severely injured or soft newborn brains in rodents. Here we report an in vivo TTC perfusion-labeling method based on osmotic opening of blood-brain-barrier with mannitol-pretreatment. This new method delineates cortical infarction correlated with the boundary of morphological cell injury, differentiates the induction or subcellular redistribution of apoptosis-related factors between viable and damaged areas, and easily determines the size of cerebral infarction in both adult and newborn mice. Using this method, we confirmed that administration of lipopolysaccharide 72 h before hypoxia-ischemia increases the damage in neonatal mouse brains, in contrast to its effect of protective preconditioning in adults. These results demonstrate a fast and inexpensive method that simplifies the task of quantifying cerebral infarction in small or severely injured brains and assists biochemical analysis of experimental cerebral ischemia.


Subject(s)
Cerebral Infarction/pathology , Coloring Agents , Mannitol/pharmacology , Staining and Labeling/methods , Tetrazolium Salts , Animals , Blood-Brain Barrier/drug effects , Disease Models, Animal , Immunoblotting , Immunohistochemistry , Male , Mice , Perfusion , Reverse Transcriptase Polymerase Chain Reaction
12.
J Cereb Blood Flow Metab ; 31(4): 1155-69, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21139628

ABSTRACT

Diffusion tensor imaging (DTI) is a powerful method to visualize white matter, but its use in patients with acute stroke remains limited because of the lack of corresponding histologic information. In this study, we addressed this issue using a hypoxia-ischemia (HI)-induced thrombotic model of stroke in adult mice. At 6, 15, and 24 hours after injury, animals were divided into three groups for (1) in vivo T2- and diffusion-weighted magnetic resonance imaging, followed by histochemistry, (2) ex vivo DTI and electron microscopy, and (3) additional biochemical or immunochemical assays. The temporal changes of diffusion anisotropy and histopathology were compared in the fimbria, internal capsule, and external capsule. We found that HI caused a rapid reduction of axial and radial diffusivities in all three axonal bundles. A large decrease in fractional anisotropy, but not in axial diffusivity per se, was associated with structural breakdown of axons. Furthermore, the decrease in radial diffusivity correlated with swelling of myelin sheaths and compression of the axoplasma. The gray matter of the hippocampus also exhibited a high level of diffusion anisotropy, and its reduction signified dendritic degeneration. Taken together, these results suggest that cross-evaluation of multiple DTI parameters may provide a fuller picture of axonal and dendritic injury in acute ischemic stroke.


Subject(s)
Brain Ischemia/pathology , Hypoxia, Brain/pathology , Intracranial Thrombosis/pathology , Stroke/pathology , Animals , Anisotropy , Axonal Transport/physiology , Axons/pathology , Brain/pathology , Brain Ischemia/complications , Diffusion Tensor Imaging , Hypoxia, Brain/complications , Immunohistochemistry , Intracranial Thrombosis/complications , Lipid Peroxidation/drug effects , Magnetic Resonance Imaging , Male , Mice , Microscopy, Electron , Oligodendroglia/drug effects , Oligodendroglia/pathology , Stroke/etiology , Time Factors
13.
Cancer Biol Ther ; 9(2): 122-33, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19923925

ABSTRACT

A common metabolic change in cancer is the acquisition of glycolytic phenotypes. Increased expression of glycolytic enzymes is considered as one contributing factor. The role of mitochondrial defects in acquisition of glycolytic phenotypes has been postulated but remains controversial. Here we show that functional defects in mitochondrial respiration could be induced by oncogenic H-Ras(Q61L) transformation, even though the mitochondrial contents or mass was not reduced in the transformed cells. First, mitochondrial respiration, as measured by mitochondrial oxygen consumption, was suppressed in NIH-3T3 cells transformed with H-Ras(Q61L). Second, oligomycin or rotenone did not reduce the cellular ATP levels in the H-Ras(Q61L) transformed cells, suggesting a diminished role of mitochondrial respiration in the cellular energy metabolism. Third, inhibition of glycolysis with iodoacetic acid reduced ATP levels at a much faster rate in H-Ras(Q61L) transformed cells than in the vector control cells. The reduction of cellular ATP levels was reversed by exogenously added pyruvate in the vector control cells but not in H-Ras(Q61L) transformed cells. Finally when compared to the HRas(Q61L) transformed cells, the vector control cells had increased resistance toward glucose deprivation. The increased resistance was dependent on mitochondrial oxidative phosphorylation since rotenone or oligomycin abolished the increased survival of the vector control cells under glucose deprivation. The results also suggest an inability of the H-Ras(Q61L) transformed cells to reactivate mitochondrial respiration under glucose deprivation. Taken together, the data suggest that mitochondrial respiration can be impaired during transformation of NIH-3T3 cells by oncogeneic H-Ras(Q61L).


Subject(s)
Cell Transformation, Neoplastic , Fibroblasts/metabolism , Genes, ras , Mitochondria/metabolism , Oncogene Protein p21(ras)/physiology , Adenosine Triphosphate/metabolism , Animals , Antimycin A/pharmacology , Electron Transport/drug effects , Energy Metabolism/drug effects , Glucose/metabolism , Glycolysis/drug effects , Iodoacetic Acid/pharmacology , Mice , Mitochondria/drug effects , Mutation, Missense , NIH 3T3 Cells/metabolism , Oligomycins/pharmacology , Oncogene Protein p21(ras)/genetics , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Point Mutation , Pyruvic Acid/pharmacology , Rotenone/pharmacology
14.
J Neurosci ; 29(27): 8669-74, 2009 Jul 08.
Article in English | MEDLINE | ID: mdl-19587273

ABSTRACT

Disruption of the integrity of the blood-brain barrier (BBB) is an important mechanism of cerebrovascular diseases, including neonatal cerebral hypoxia-ischemia (HI). Although both tissue-type plasminogen activator (tPA) and matrix metalloproteinase-9 (MMP-9) can produce BBB damage, their relationship in neonatal cerebral HI is unclear. Here we use a rodent model to test whether the plasminogen activator (PA) system is critical for MMP-9 activation and HI-induced brain injury in newborns. To test this hypothesis, we examined the therapeutic effect of intracerebroventricular injection of plasminogen activator inhibitor-1 (PAI-1) in rat pups subjected to unilateral carotid artery occlusion and systemic hypoxia. We found that the injection of PAI-1 greatly reduced the activity of both tPA and urokinase-type plasminogen activator after HI. It also blocked HI-induced MMP-9 activation and BBB permeability at 24 h of recovery. Furthermore, magnetic resonance imaging and histological analysis showed the PAI-1 treatment reduced brain edema, axonal degeneration, and cortical cell death at 24-48 h of recovery. Finally, the PAI-1 therapy provided a dose-dependent decrease of brain tissue loss at 7 d of recovery, with the therapeutic window at 4 h after the HI insult. Together, these results suggest that the brain PA system plays a pivotal role in neonatal cerebral HI and may be a promising therapeutic target in infants suffering hypoxic-ischemic encephalopathy.


Subject(s)
Hypoxia-Ischemia, Brain/prevention & control , Hypoxia-Ischemia, Brain/physiopathology , Plasminogen Activator Inhibitor 1/administration & dosage , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Humans , Hypoxia-Ischemia, Brain/enzymology , Injections, Intraventricular , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase Inhibitors , Protease Inhibitors/administration & dosage , Rats , Rats, Wistar , Serine Proteinase Inhibitors/administration & dosage
15.
Int J Cancer ; 124(7): 1545-51, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19089921

ABSTRACT

The enzyme 15-lipoxygenase-2 (15-LOX-2) utilizes arachidonic acid, a polyunsaturated fatty acid, to synthesize 15(S)-hydroxyeicosatetraenoic acid. Abundantly expressed in normal prostate epithelium but frequently suppressed in the cancerous tissues, 15-LOX-2 has been suggested as a functional suppressor of prostate cancer, but the mechanism(s) involved remains unknown. To study the functional role of 15-LOX-2 in prostate cancer, we expressed 15-LOX-2 as a fusion protein with GFP in DU145 and PC-3 cells and found that 15-LOX-2 increased cell cycle arrest at G0/G1 phase. When injected into athymic nu/nu mice, prostate cancer cells with 15-LOX-2 expression could still form palpable tumors without significant changes in tumorigenicity. But, the tumors with 15-LOX-2 expression grew significantly slower than those derived from vector controls and were kept dormant for a long period of time. Histological evaluation revealed an increase in cell death in tumors derived from prostate cancer cells with 15-LOX-2 expression, while in vitro cell culture conditions, no such increase in apoptosis was observed. Further studies found that the expression of vascular endothelial growth factor A (VEGF-A) was significantly reduced in prostate cancer cells with 15-LOX-2 expression restored. Our studies suggest that 15-LOX-2 suppresses VEGF gene expression and sustains tumor dormancy in prostate cancer. Loss of 15-LOX-2 functionalities, therefore, represents a key step for prostate cancer cells to exit from dormancy and embark on malignant progression in vivo.


Subject(s)
Arachidonate 15-Lipoxygenase/metabolism , Gene Expression Regulation, Neoplastic/physiology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Vascular Endothelial Growth Factor A/metabolism , Animals , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression , Humans , Immunohistochemistry , Male , Mice , Prostatic Neoplasms/genetics , Recombinant Fusion Proteins , Transfection , Vascular Endothelial Growth Factor A/genetics
16.
Cancer Res ; 68(15): 6396-406, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18676865

ABSTRACT

Our previous study revealed that Vav3 oncogene is overexpressed in human prostate cancer, activates androgen receptor (AR), and stimulates growth in prostate cancer cells. The purpose of this study is to further determine the potential role of Vav3 in prostate cancer development in genetically engineered mouse model. We generated Vav3 transgenic mice by targeted overexpression of a constitutive active Vav3 in the prostatic epithelium. We found that overexpression of Vav3 led to development of mouse prostatic intraepithelial neoplasia and prostate cancer at the age of as early as 3 months. The AR signaling axis and phosphatidylinositol 3-kinase-Akt signaling were elevated in the prostate glands of Vav3 transgenic mice. In addition to prostate cancer, Vav3 transgenic mice developed significant nonbacterial chronic prostatitis in the prostate gland with notable infiltration of lymphomononuclear cells (monocytes, lymphocytes, and plasma cells), which was associated with elevated incidence of prostate cancer. DNA microarray and signaling pathway analysis revealed that the top diseases and disorders were inflammatory diseases and cancer of the prostate gland in Vav3 transgenic mice. In vitro analysis showed that overexpression of Vav3 in prostate cancer cells enhanced nuclear factor-kappaB (NF-kappaB) activity, implicating an underlying mechanism of innate inflammatory response induced by elevated Vav3 activity. These data showed that Vav3 overexpression in the prostate epithelium enhanced both the AR signaling axis and NF-kappaB-mediated pathway, which potentially contributed to the development of nonbacterial prostatitis and prostate cancer.


Subject(s)
Prostate/metabolism , Prostatic Neoplasms/genetics , Prostatitis/genetics , Proto-Oncogene Proteins c-vav/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Epithelium/metabolism , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/metabolism , Prostatitis/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
17.
Cancer Res ; 68(1): 115-21, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18172303

ABSTRACT

Thromboxane A(2) (TxA(2)) is a prostanoid formed by thromboxane synthase using the cyclooxygenase product prostaglandin H(2) as the substrate. Previously, increased expression of thromboxane synthase was found in prostate tumors, and tumor cell motility was attenuated by inhibitors of thromboxane synthase. This study was undertaken to elucidate how tumor motility is regulated by TxA(2). Here, we report that human prostate cancer cells express functional receptors for TxA(2) (TP). Ligand binding assay found that PC-3 cells binded to SQ29548, a high-affinity TP antagonist, in a saturable manner with K(d) of 3.64 nmol/L and B(max) of 120.4 fmol per million cells. Treatment of PC-3 cells by U46619, a TP agonist, induced PC-3 cell contraction, which was blocked by pretreatment with the TP antagonist SQ29548 or pinane TxA(2). The migration of prostate cancer cells was significantly inhibited either by sustained activation of TP or by blockade of TP activation, suggesting that TP activation must be tightly controlled during cell migration. Further studies found that small GTPase RhoA was activated by TP activation, and pretreatment of PC-3 cells with Y27632, a Rho kinase (ROCK) inhibitor, blocked U46619-induced cell contraction. A dominant-negative mutant of RhoA also blocked U46619-induced cell contraction. Taken together, the data suggest that TPs are expressed in prostate cancer and activation of TPs regulates prostate cancer cell motility and cytoskeleton reorganization through activation of Rho.


Subject(s)
Carcinoma/pathology , Cell Movement , Prostatic Neoplasms/pathology , Receptors, Thromboxane A2, Prostaglandin H2/physiology , rhoA GTP-Binding Protein/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Amides/pharmacology , Bridged Bicyclo Compounds, Heterocyclic , Carcinoma/chemistry , Carcinoma/metabolism , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Fatty Acids, Unsaturated , Humans , Hydrazines/pharmacology , Ligands , Male , Prostatic Neoplasms/chemistry , Prostatic Neoplasms/metabolism , Pyridines/pharmacology , Receptors, Thromboxane A2, Prostaglandin H2/analysis , Receptors, Thromboxane A2, Prostaglandin H2/drug effects , rhoA GTP-Binding Protein/analysis , rhoA GTP-Binding Protein/antagonists & inhibitors
18.
Yi Chuan Xue Bao ; 29(10): 903-6, 2002 Oct.
Article in Chinese | MEDLINE | ID: mdl-12561476

ABSTRACT

Southern corn rust (SCR) is a destructive disease in maize. The inbred line Qi319 is highly resistant to southern corn rust. The inheritance of resistance to southern rust in Qi319 was investigated. Five F1 hybrids were derived from Qi319 crossed with five susceptible inbred lines respectively. The F2 generations were produced by F1 self-pollinated and BC1 F1 generations were abtained by backcrossing F1 with the susceptible parents. Inoculation of the P1, P2, F1 s and 10 individuals of F2, BC1 F1 were completed with the southern corn rust pathogen and showed that all of the 10 F1 s were resistant, the all 5 F2 populations segregated in a ratio of 3R:1S, and all of the 5 BC1 F1 populations segregated in a ratio of 1R:1S. Therefore, it is considered that Qi319 carries one dominant gene for resistance to southern corn rust.


Subject(s)
Basidiomycota/growth & development , Zea mays/genetics , Genes, Dominant/genetics , Immunity, Innate/genetics , Inbreeding , Models, Genetic , Plant Diseases/genetics , Plant Diseases/microbiology , Zea mays/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...