Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Stroke ; 54(3): 857-867, 2023 03.
Article in English | MEDLINE | ID: mdl-36734233

ABSTRACT

BACKGROUND: Stroke is a leading cause of morbidity and mortality worldwide. Neuroinflammation plays a key role in acute brain injury of ischemic stroke. MicroRNA-210 (miR210) is the master hypoxamir and regulates microglial activation and inflammation in a variety of diseases. In this study, we uncovered the mechanism of miR210 in orchestrating ischemic stroke-induced neuroinflammation through repression of TET2 (ten-eleven translocation methylcytosine dioxygenase 2) in the adult mouse brain. METHODS: Ischemic stroke was induced in adult WT (wild type) or miR210 KO (miR210 deficient) mice by transient intraluminal middle cerebral artery occlusion. Injection of TET2 silencing RNA or miR210 complementary locked nucleic acid oligonucleotides, or miR210 KO mice were used to validate miR210-TET2 axis and its role in ischemic brain injury. Furthermore, the effect of TET2 overexpression on miR210-stimulated proinflammatory cytokines was examined in BV2 microglia. Post assays included magnetic resonance imaging scan for brain infarct size; neurobehavioral tests, reverse transcription-quantitative polymerase chain reaction, and Western blot for miR210; and TET2 levels, flow cytometry, and ELISA for neuroinflammation in the brain after stroke or microglia in vitro. RESULTS: miR210 injection significantly reduced TET2 protein abundance in the brain, while miR210 complementary locked nucleic acid oligonucleotides or miR210 KO preserved TET2 regardless of ischemic brain injury. TET2 knockdown reversed the protective effects of miR210 inhibition or miR210 KO on ischemic stroke-induced brain infarct size and neurobehavioral deficits. Moreover, flow cytometry and ELISA assays showed that TET2 knockdown also significantly dampened the anti-inflammatory effect of miR210 inhibition on microglial activation and IL (interleukin)-6 release after stroke. In addition, overexpression of TET2 in BV2 microglia counteracted miR210-induced increase in cytokines. CONCLUSIONS: miR210 inhibition reduced ischemic stroke-induced neuroinflammatory response via repression of TET2 in the adult mouse brain, suggesting that miR210 is a potential treatment target for acute brain injury after ischemic stroke.


Subject(s)
Brain Injuries , Brain Ischemia , Dioxygenases , Ischemic Stroke , MicroRNAs , Animals , Mice , Brain Injuries/pathology , Brain Ischemia/pathology , Cytokines/metabolism , Infarction/pathology , Infarction, Middle Cerebral Artery/pathology , Inflammation/pathology , Interleukin-6/metabolism , Ischemic Stroke/pathology , Mice, Inbred C57BL , Microglia/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Neuroinflammatory Diseases
2.
BMC Vet Res ; 19(1): 131, 2023 Aug 23.
Article in English | MEDLINE | ID: mdl-37612662

ABSTRACT

BACKGROUND: Chronic kidney disease (CKD) is a common cause of morbidity and mortality in captive wildlife species. However, CKD has been rarely documented in giant pandas. CASE PRESENTATION: The following report describes a case of an eight-year-old female giant panda showing clinical signs of epistaxis, bloody diarrhea, polyuria, azotemia and anemia. The animal died despite of supportive treatments. Necropsy was performed. Grossly, both kidneys were shrunken and scarred with pallor. Subcutis edema and petechia on the epicardium of the heart were observed. The tissue samples were made into paraffin sections and stained by H.E and special staining including Periodic Acid-Schiff (PAS), von Kossa, Masson's trichrome, Phosphotungstic acid-hematoxylin (PTAH), and Congo red. Histopathology examination revealed severe chronic tubulointerstitial nephritis with marked interstitial fibrosis, glomerulosclerosis, tubular atrophy and calcification in kidneys, and acute necrotizing hemorrhagic myocarditis with calcification in heart. Other lesions included intestinal hemorrhage, hepatic fatty degeneration and necrosis with hemosiderin, and splenic hemosiderin. CONCLUSIONS: In summary, chronic kidney disease was finally diagnosed based on the association of clinical, gross, and histopathological findings. Heart failure secondary to CKD is the leading cause of death in this giant panda. The potential cause of CKD in this animal is possibly due to long term and uncontrolled hypertension. Blood pressure monitoring is essential in establishing the diagnosis and management of hypertension in giant panda.


Subject(s)
Hypertension , Renal Insufficiency, Chronic , Ursidae , Animals , Female , Hemosiderin , Renal Insufficiency, Chronic/veterinary , Kidney , Hypertension/veterinary
3.
J Neuroinflammation ; 18(1): 6, 2021 Jan 05.
Article in English | MEDLINE | ID: mdl-33402183

ABSTRACT

BACKGROUND: Neonatal hypoxic-ischemic (HI) brain injury is a leading cause of acute mortality and chronic disability in newborns. Our previous studies demonstrated that HI insult significantly increased microRNA-210 (miR-210) in the brain of rat pups and inhibition of brain endogenous miR-210 by its inhibitor (LNA) provided neuroprotective effect in HI-induced brain injury. However, the molecular mechanisms underpinning this neuroprotection remain unclear. METHODS: We made a neonatal HI brain injury model in mouse pups of postnatal day 7 to uncover the mechanism of miR-210 in targeting the ten eleven translocation (TET) methylcytosine dioxygenase 2 that is a transcriptional suppressor of pro-inflammatory cytokine genes in the neonatal brain. TET2 silencing RNA was used to evaluate the role of TET2 in the neonatal HI-induced pro-inflammatory response and brain injury. MiR-210 mimic and inhibitor (LNA) were delivered into the brain of mouse pups to study the regulation of miR-210 on the expression of TET2. Luciferase reporter gene assay was performed to validate the direct binding of miR-210 to the 3' untranslated region of the TET2 transcript. Furthermore, BV2 mouse microglia cell line was employed to confirm the role of miR-210-TET2 axis in regulating pro-inflammatory response in microglia. Post-assays included chromatin immunoprecipitation (ChIP) assay, co-immunoprecipitation, RT-PCR, brain infarct assay, and neurobehavioral test. Student's t test or one-way ANOVA was used for statistical analysis. RESULTS: HI insult significantly upregulated miR-210, downregulated TET2 protein abundance, and increased NF-κB subunit p65 acetylation level and its DNA binding capacity to the interleukin 1 beta (IL-1ß) promoter in the brain of mouse pups. Inhibition of miR-210 rescued TET2 protein level from HI insult and miR-210 mimic decreased TET2 protein level in the brain of mouse pups, suggesting that TET2 is a functional target of miR-210. The co-immunoprecipitation was performed to reveal the role of TET2 in HI-induced inflammatory response in the neonatal brain. The result showed that TET2 interacted with NF-κB subunit p65 and histone deacetylase 3 (HDAC3), a co-repressor of gene transcription. Furthermore, TET2 knockdown increased transcriptional activity of acetyl-p65 on IL-1ß gene in the neonatal brain and enhanced HI-induced upregulation of acetyl-p65 level and pro-inflammatory cytokine expression. Of importance, TET2 knockdown exacerbated brain infarct size and neurological deficits and counteracted the neuroprotective effect of miR-210 inhibition. Finally, the in vitro results demonstrated that the miR-210-TET2 axis regulated pro-inflammatory response in BV2 mouse microglia cell line. CONCLUSIONS: The miR-210-TET2 axis regulates pro-inflammatory cytokine expression in microglia, contributing to neonatal HI brain injury.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , Down-Regulation/physiology , Hypoxia-Ischemia, Brain/metabolism , Inflammation Mediators/metabolism , MicroRNAs/biosynthesis , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/biosynthesis , Animals , Animals, Newborn , Cell Line , Dioxygenases , Female , Hypoxia-Ischemia, Brain/pathology , Male , Mice
4.
Ecotoxicol Environ Saf ; 219: 112363, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34087735

ABSTRACT

Persistent organic pollutant (POPs) contamination was analyzed in samples collected from wild and captive giant pandas to characterize seasonal variation in concentrations of POPs and possible sources. POP concentrations in bamboo and fecal samples collected from captive pandas showed significant fluctuations compared with those collected from wild pandas in each season. The highest polychlorinated biphenyl (PCB) and organochlorine pesticide (OCP) concentrations were 1380 pg g-1 dw and 3140 pg g-1 dw, respectively, which were observed in captive bamboo samples in the summer. PCBs varied seasonally, whereas OCPs did not show apparent seasonal variation. Based on the seasonal variability, component analysis, and the positive matrix factorization results, we determined that the secondary volatilization of POPs during periods of high temperatures was the leading cause of the exposure of pandas to pollutants (45%), and atmospheric transport played a crucial role in the secondary distribution of pollutants in panda food. The other two sources of pollution were historical residues transmitted over long distances to protected areas (28%), as well as UP-POPs and new inputs from agricultural activities (27%). The concentrations of pollutants in bamboo shoots were significantly lower than those in bamboo. Therefore, bamboo shoots should be incorporated into the diet of captive pandas in the spring to reduce their exposure to pollutants. The absorption capacity of pollutants associated with the consumption of bamboo shoots was significantly lower than that associated with the consumption of bamboo. The diet of young captive pandas in the summer should also be managed with caution given their slightly stronger ability to absorb pollutants.


Subject(s)
Environmental Exposure/statistics & numerical data , Environmental Pollutants/metabolism , Environmental Pollution/statistics & numerical data , Ursidae/metabolism , Animals , Diet , Environmental Monitoring , Polychlorinated Biphenyls , Seasons
5.
Int J Mol Sci ; 22(16)2021 Aug 20.
Article in English | MEDLINE | ID: mdl-34445671

ABSTRACT

C-type natriuretic peptide (CNP) is an important vascular regulator that is present in the brain. Our previous study demonstrated the innate neuroprotectant role of CNP in the neonatal brain after hypoxic-ischemic (HI) insults. In this study, we further explored the role of CNP in cerebrovascular pathology using both in vivo and in vitro models. In a neonatal mouse HI brain injury model, we found that intracerebroventricular administration of recombinant CNP dose-dependently reduces brain infarct size. CNP significantly decreases brain edema and immunoglobulin G (IgG) extravasation into the brain tissue, suggesting a vasculoprotective effect of CNP. Moreover, in primary brain microvascular endothelial cells (BMECs), CNP dose-dependently protects BMEC survival and monolayer integrity against oxygen-glucose deprivation (OGD). The vasculoprotective effect of CNP is mediated by its innate receptors NPR2 and NPR3, in that inhibition of either NPR2 or NPR3 counteracts the protective effect of CNP on IgG leakage after HI insult and BMEC survival under OGD. Of importance, CNP significantly ameliorates brain atrophy and improves neurological deficits after HI insults. Altogether, the present study indicates that recombinant CNP exerts vascular protection in neonatal HI brain injury via its innate receptors, suggesting a potential therapeutic target for the treatment of neonatal HI brain injury.


Subject(s)
Hypoxia-Ischemia, Brain/pathology , Natriuretic Peptide, C-Type/pharmacology , Vascular System Injuries/prevention & control , Animals , Animals, Newborn , Brain/metabolism , Brain Edema/pathology , Brain Infarction/metabolism , Brain Injuries/pathology , Cells, Cultured , Endothelial Cells/metabolism , Female , Hypoxia-Ischemia, Brain/metabolism , Infusions, Intraventricular , Male , Mice , Natriuretic Peptide, C-Type/metabolism , Natriuretic Peptide, C-Type/physiology , Neuroprotective Agents , Vascular System Injuries/metabolism
6.
Am J Physiol Cell Physiol ; 317(1): C3-C19, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30840494

ABSTRACT

MicroRNAs (miRNAs) are a class of highly conserved non-coding RNAs with 21-25 nucleotides in length and play an important role in regulating gene expression at the posttranscriptional level via base-paring with complementary sequences of the 3'-untranslated region of the target gene mRNA, leading to either transcript degradation or translation inhibition. Brain-enriched miRNAs act as versatile regulators of brain development and function, including neural lineage and subtype determination, neurogenesis, synapse formation and plasticity, neural stem cell proliferation and differentiation, and responses to insults. Herein, we summarize the current knowledge regarding the role of miRNAs in brain development and cerebrovascular pathophysiology. We review recent progress of the miRNA-based mechanisms in neuronal and cerebrovascular development as well as their role in hypoxic-ischemic brain injury. These findings hold great promise, not just for deeper understanding of basic brain biology but also for building new therapeutic strategies for prevention and treatment of pathologies such as cerebral ischemia.


Subject(s)
Brain/blood supply , Brain/metabolism , Cerebrovascular Disorders/metabolism , MicroRNAs/metabolism , Neovascularization, Pathologic , Neovascularization, Physiologic , Neurogenesis , Neurons/metabolism , Animals , Brain/growth & development , Cerebrovascular Disorders/genetics , Cerebrovascular Disorders/physiopathology , Gene Expression Regulation, Developmental , Humans , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/physiopathology , MicroRNAs/genetics , Signal Transduction
7.
Brain Inj ; 33(6): 712-716, 2019.
Article in English | MEDLINE | ID: mdl-30335519

ABSTRACT

Aim: In this work, we systematically explored the physiological functions of astrocytes and their roles following ischemic stroke, additionally, the potential therapy strategy targeting the astrocytes was also discussed. Methods: This work searched the PubMed database (including MEDLINE) until 14 Feb 2018, and furthermore, the studies were identified through cross-referencing and by consulting the experts in this field. Results: This study indicated that the astrocytes can not only play harmful roles following ischemic stroke through release of inflammatory factors and formation of glial scar but also have protective effects through quenching glutamate excitotoxicity and maintaining the clearance function of glymphatic system in brain. Conclusion: Owing to their important roles in physiological functions of brain and in the pathological conditions following ischemic stroke, the astrocytes might be a potential but promising therapeutic target for treating the ischemic stroke in the future.


Subject(s)
Astrocytes/pathology , Brain Ischemia/pathology , Brain Ischemia/therapy , Stroke/pathology , Humans , Neuroprotection , Stroke/therapy
9.
Ecotoxicol Environ Saf ; 162: 218-224, 2018 Oct 30.
Article in English | MEDLINE | ID: mdl-29990734

ABSTRACT

Captive pandas are exposed to higher concentrations of environmental toxins in their food source and from atmospheric pollution than wild pandas. Moreover, the Qinling panda subspecies had significantly higher concentrations of toxic chemicals in its feces. To determine whether these toxicants also accumulate in panda's blood and impair its health, concentrations of persistent organic pollutants (POPs) and heavy metals were measured in blood samples. Four heavy metals (As, Cd, Cr and Pb), PCDD/Fs and PCBs were detected in blood drawn from captive Qinling pandas. Time spent in captivity was a better predictor of toxicant concentration accumulation than was panda age. More than 50% of the studied pandas were outside the normal levels for 11 health parameters, and five (ALT, LDH, Ca, Cl, TB) of the 11 parameters classified as abnormal were correlated with blood pollutant concentrations. The proportion of live sperm was significantly lower and the aberrance ratio of sperm was significantly greater for captive pandas than for wild ones. A short-term solution to reduce the health impacts of pollution and toxicant exposure of Qinling pandas is to relocate breeding centers to less contaminated areas and to strictly control the quality of their food provided. A longer term solution depends on improving air quality by reducing toxic emissions.


Subject(s)
Environmental Pollutants/toxicity , Kidney/drug effects , Liver/drug effects , Metals, Heavy/toxicity , Spermatozoa/drug effects , Ursidae/physiology , Animals , Environmental Pollutants/blood , Environmental Pollution , Male , Metals, Heavy/blood , Polychlorinated Biphenyls/blood , Polychlorinated Biphenyls/toxicity , Semen Analysis/veterinary
10.
Int J Mol Sci ; 19(8)2018 Aug 17.
Article in English | MEDLINE | ID: mdl-30126083

ABSTRACT

Strokes are one of the leading causes of mortality and chronic morbidity in the world, yet with only limited successful interventions available at present. Our previous studies revealed the potential role of the glucocorticoid receptor (GR) in the pathogenesis of neonatal hypoxic-ischemic encephalopathy (HIE). In the present study, we investigate the effect of GR knockdown on acute ischemic brain injuries in a model of focal cerebral ischemia induced by middle cerebral artery occlusion (MCAO) in adult male CD1 mice. GR siRNAs and the negative control were administered via intracerebroventricular (i.c.v.) injection 48 h prior to MCAO. The cerebral infarction volume and neurobehavioral deficits were determined 48 h after MCAO. RT-qPCR was employed to assess the inflammation-related gene expression profiles in the brain before and after MCAO. Western Blotting was used to evaluate the expression levels of GR, the mineralocorticoid receptor (MR) and the brain-derived neurotrophic factor/tropomyosin receptor kinase B (BDNF/TrkB) signaling. The siRNAs treatment decreased GR, but not MR, protein expression, and significantly enhanced expression levels of pro-inflammatory cytokines (IL-6, IL-1ß, and TNF-α) in the brain. Of interest, GR knockdown suppressed BDNF/TrkB signaling in adult mice brains. Importantly, GR siRNA pretreatment significantly increased the infarction size and exacerbated the neurobehavioral deficits induced by MCAO in comparison to the control group. Thus, the present study demonstrates the important role of GR in the regulation of the inflammatory responses and neurotrophic BDNF/TrkB signaling pathway in acute ischemic brain injuries in adult mice, revealing a new insight into the pathogenesis and therapeutic potential in acute ischemic strokes.


Subject(s)
Brain/pathology , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Receptors, Glucocorticoid/genetics , Animals , Brain/metabolism , Gene Knockdown Techniques , Infarction, Middle Cerebral Artery/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice , RNA Interference , RNA, Small Interfering/genetics , Receptors, Glucocorticoid/metabolism , Signal Transduction
11.
Ecol Appl ; 27(2): 343-348, 2017 03.
Article in English | MEDLINE | ID: mdl-28039891

ABSTRACT

The giant panda (Ailuropoda melanoleuca) is one of the most endangered animals in the world, and it is recognized worldwide as a symbol for conservation. A previous study showed that wild and captive pandas, especially those of the Qinling subspecies, were exposed to toxicants in their diet of bamboo; the ultimate origin of these toxicants is unknown. Here we show that atmospheric deposition is the most likely origin of heavy metals and persistent organic pollutants (POPs) in the diets of captive and wild Qinling pandas. Average atmospheric deposition was 199, 115, and 49 g·m-2 ·yr-1 in the center of Xi'an City, at China's Shaanxi Wild Animal Research Center (SWARC), and at Foping National Nature Reserve (FNNR), respectively. Atmospheric deposition of heavy metals (As, Cd, Cr, Pb, Hg, Co, Cu, Zn, Mn, and Ni) and POPs was highest at Xi'an City, intermediate at SWARC, and lowest at FNNR. Soil concentrations of the aforementioned heavy metals other than As and Zn also were significantly higher at SWARC than at FNNR. Efforts to conserve Qinling pandas may be compromised by air pollution attendant to China's economic development. Improvement of air quality and reductions of toxic emissions are urgently required to protect China's iconic species.


Subject(s)
Environmental Monitoring , Environmental Pollutants/analysis , Metals, Heavy/analysis , Ursidae/metabolism , Animals , Diet
12.
Proc Natl Acad Sci U S A ; 111(11): E1035-42, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24591593

ABSTRACT

Humans with ALS and transgenic rodents expressing ALS-associated superoxide dismutase (SOD1) mutations develop spontaneous blood-spinal cord barrier (BSCB) breakdown, causing microvascular spinal-cord lesions. The role of BSCB breakdown in ALS disease pathogenesis in humans and mice remains, however, unclear, although chronic blood-brain barrier opening has been shown to facilitate accumulation of toxic blood-derived products in the central nervous system, resulting in secondary neurodegenerative changes. By repairing the BSCB and/or removing the BSCB-derived injurious stimuli, we now identify that accumulation of blood-derived neurotoxic hemoglobin and iron in the spinal cord leads to early motor-neuron degeneration in SOD1(G93A) mice at least in part through iron-dependent oxidant stress. Using spontaneous or warfarin-accelerated microvascular lesions, motor-neuron dysfunction and injury were found to be proportional to the degree of BSCB disruption at early disease stages in SOD1(G93A) mice. Early treatment with an activated protein C analog restored BSCB integrity that developed from spontaneous or warfarin-accelerated microvascular lesions in SOD1(G93A) mice and eliminated neurotoxic hemoglobin and iron deposits. Restoration of BSCB integrity delayed onset of motor-neuron impairment and degeneration. Early chelation of blood-derived iron and antioxidant treatment mitigated early motor-neuronal injury. Our data suggest that BSCB breakdown contributes to early motor-neuron degeneration in ALS mice and that restoring BSCB integrity during an early disease phase retards the disease process.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Blood-Nerve Barrier/pathology , Motor Neurons/pathology , Nerve Degeneration/physiopathology , Spinal Cord/pathology , Animals , Blood-Nerve Barrier/physiology , Ferrocyanides , Humans , Immunoblotting , In Situ Nick-End Labeling , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Motor Neurons/physiology , Point Mutation/genetics , Protein C/metabolism , Real-Time Polymerase Chain Reaction , Rotarod Performance Test , Spinal Cord/physiology , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism , Warfarin
13.
Ecotoxicol Environ Saf ; 143: 72-79, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28505482

ABSTRACT

Chemical elements exposure of endangered golden takins (Budorcas taxicolor bedfordi) living in the Qinling Mountains and in a captive breeding center was assessed by analyzing fecal samples. Concentrations of As, Co, Cr, Cu, Ni and Se were significantly higher in the feces of captive golden takins than the wild. There was no significant difference in the fecal concentrations of Cd, Mn, Hg, Pb or Zn for wild and captive animals. The element concentration of fecal samples collected from captive animals varied seasonally, with concentrations being lowest in spring and highest in winter and/or autumn. The food provided to captive animals varied both in the composition and the concentration of element present. Consumptions of feedstuff and additional foods such as D. sanguinalis and A. mangostanus for the captive golden takins were identified as the possible sources of chemical element exposure. The estimations of dietary intake of most elements by captive takins were below the oral reference dose, except for As and Pb, indicating that As and Pb were the key components which contributed to the potential non-carcinogenic risk for captive golden takins. In conclusion, captive golden takins were exposed to higher concentrations of chemical elements compared with the wild, which were likely due to their dietary difference. Conservation efforts of captive golden takin are potentially compromised by the elevated chemical element exposure and effort should focus on providing uncontaminated food for captive animals.


Subject(s)
Conservation of Natural Resources , Endangered Species , Environmental Monitoring , Feces/chemistry , Metals, Heavy/analysis , Ruminants/growth & development , Altitude , Animal Feed/standards , Animals , China , Metals, Heavy/metabolism , Ruminants/metabolism , Seasons
14.
Int J Mol Sci ; 18(7)2017 Jun 24.
Article in English | MEDLINE | ID: mdl-28672801

ABSTRACT

Cerebral edema, primarily caused by disruption of the blood-brain barrier (BBB), is one of the serious complications associated with brain injury in neonatal hypoxic-ischemic encephalopathy (HIE). Our recent study demonstrated that the hypoxic-ischemic (HI) treatment significantly increased microRNA-210 (miR-210) in the neonatal rat brain and inhibition of miR-210 provided neuroprotection in neonatal HI brain injury. The present study aims to determine the role of miR-210 in the regulation of BBB integrity in the developing brain. miR-210 mimic was administered via intracerebroventricular injection (i.c.v.) into the brain of rat pups. Forty-eight hours after the injection, a modified Rice-Vannucci model was conducted to produce HI brain injury. Post-assays included cerebral edema analysis, western blotting, and immunofluorescence staining for serum immunoglobulin G (IgG) leakage. The results showed that miR-210 mimic exacerbated cerebral edema and IgG leakage into the brain parenchyma. In contrast, inhibition of miR-210 with its complementary locked nucleic acid oligonucleotides (miR-210-LNA) significantly reduced cerebral edema and IgG leakage. These findings suggest that miR-210 negatively regulates BBB integrity i n the neonatal brain. Mechanistically, the seed sequences of miR-210 were identified complementary to the 3' untranslated region (3' UTR) of the mRNA transcripts of tight junction protein occludin and adherens junction protein ß-catenin, indicating downstream targets of miR-210. This was further validated by in vivo data showing that miR-210 mimic significantly reduced the expression of these junction proteins in rat pup brains. Of importance, miR-210-LNA preserved the expression of junction proteins occludin and ß-catenin from neonatal HI insult. Altogether, the present study reveals a novel mechanism of miR-210 in impairing BBB integrity that contributes to cerebral edema formation after neonatal HI insult, and provides new insights in miR-210-LNA mediated neuroprotection in neonatal HI brain injury.


Subject(s)
Blood-Brain Barrier/metabolism , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/metabolism , Intercellular Junctions/genetics , Intercellular Junctions/metabolism , MicroRNAs/genetics , Animals , Animals, Newborn , Biomarkers , Brain Edema/genetics , Brain Edema/metabolism , Brain Edema/pathology , Disease Models, Animal , Female , Gene Expression Regulation , Hypoxia-Ischemia, Brain/pathology , Immunoglobulin G/metabolism , Male , Occludin/metabolism , Permeability , RNA Interference , Rats , beta Catenin/metabolism
15.
J Mol Cell Cardiol ; 91: 160-71, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26779948

ABSTRACT

Large studies in humans and animals have demonstrated a clear association of an adverse intrauterine environment with an increased risk of cardiovascular disease later in life. Yet mechanisms remain largely elusive. The present study tested the hypothesis that gestational hypoxia leads to promoter hypermethylation and epigenetic repression of the glucocorticoid receptor (GR) gene in the developing heart, resulting in increased heart susceptibility to ischemia and reperfusion injury in offspring. Hypoxic treatment of pregnant rats from day 15 to 21 of gestation resulted in a significant decrease of GR exon 14, 15, 16, and 17 transcripts, leading to down-regulation of GR mRNA and protein in the fetal heart. Functional cAMP-response elements (CREs) at -4408 and -3896 and Sp1 binding sites at -3425 and -3034 were identified at GR untranslated exon 1 promoters. Hypoxia significantly increased CpG methylation at the CREs and Sp1 binding sites and decreased transcription factor binding to GR exon 1 promoter, accounting for the repression of the GR gene in the developing heart. Of importance, treatment of newborn pups with 5-aza-2'-deoxycytidine reversed hypoxia-induced promoter methylation, restored GR expression and prevented hypoxia-mediated increase in ischemia and reperfusion injury of the heart in offspring. The findings demonstrate a novel mechanism of epigenetic repression of the GR gene in fetal stress-mediated programming of ischemic-sensitive phenotype in the heart.


Subject(s)
Epigenesis, Genetic , Hypoxia/genetics , Myocardial Reperfusion Injury/genetics , Oxygen/pharmacology , Receptors, Glucocorticoid/genetics , Sp1 Transcription Factor/genetics , Animals , Animals, Newborn , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Binding Sites , DNA Methylation/drug effects , Decitabine , Exons , Female , Hypoxia/drug therapy , Hypoxia/metabolism , Hypoxia/pathology , Male , Maternal Exposure , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Phenotype , Pregnancy , Promoter Regions, Genetic , Protein Binding , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/metabolism , Response Elements , Sp1 Transcription Factor/metabolism
16.
Neurobiol Dis ; 89: 202-12, 2016 May.
Article in English | MEDLINE | ID: mdl-26875527

ABSTRACT

Perinatal hypoxic-ischemic encephalopathy (HIE) is associated with high neonatal mortality and severe long-term neurologic morbidity. Yet the mechanisms of brain injury in infants with HIE remain largely elusive. The present study determined a novel mechanism of microRNA-210 (miR-210) in silencing endogenous neuroprotection and increasing hypoxic-ischemic brain injury in neonatal rats. The study further revealed a potential therapeutic effect of miR-210 inhibition using complementary locked nucleic acid oligonucleotides (miR-210-LNA) in 10-day-old neonatal rats in the Rice-Vannucci model. The underlying mechanisms were investigated with intracerebroventricular injection (i.c.v) of miR-210 mimic, miR-210-LNA, glucocorticoid receptor (GR) agonist and antagonist. Luciferase reporter gene assay was conducted for identification of miR-210 targeting GR 3'untranslated region. The results showed that the HI treatment significantly increased miR-210 levels in the brain, and miR-210 mimic significantly decreased GR protein abundance and exacerbated HI brain injury in the pups. MiR-210-LNA administration via i.c.v. 4h after the HI insult significantly decreased brain miR-210 levels, increased GR protein abundance, reduced HI-induced neuronal death and brain infarct size, and improved long-term neurological function recovery. Of importance, the intranasal delivery of miR-210-LNA 4h after the HI insult produced similar effects in decreasing HI-induced neonatal brain injury and improving neurological function later in life. Altogether, the present study provides evidence of a novel mechanism of miR-210 in a neonatal HI brain injury model, and suggests a potential therapeutic approach of miR-210 inhibition in the treatment of neonatal HIE.


Subject(s)
Brain/metabolism , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/prevention & control , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Neuroprotective Agents/administration & dosage , Oligonucleotides/administration & dosage , 3' Untranslated Regions , Animals , Animals, Newborn , Rats , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism
17.
J Neurosci Res ; 93(1): 94-103, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25196554

ABSTRACT

Blood-brain barrier (BBB) disruption and consequent edema formation contribute to the development of early brain injury following subarachnoid hemorrhage (SAH). Various cerebrovascular insults result in increased platelet-derived growth factor receptor (PDGFR)-α stimulation, which has been linked to BBB breakdown and edema formation. This study examines whether imatinib, a PDGFR inhibitor, can preserve BBB integrity in a rat endovascular perforation SAH model. Imatinib (40 or 120 mg/kg) or a vehicle was administered intraperitoneally at 1 hr after SAH induction. BBB leakage, brain edema, and neurological deficits were evaluated. Total and phosphorylated protein expressions of PDGFR-α, c-Src, c-Jun N-terminal kinase (JNK), and c-Jun were measured, and enzymatic activities of matrix metalloproteinase (MMP)-2 and MMP-9 were determined in the injured brain. Imatinib treatment significantly ameliorated BBB leakage and edema formation 24 hr after SAH, which was paralleled by improved neurological functions. Decreased brain expressions of phosphorylated PDGFR-α, c-Src, JNK, and c-Jun as well as reduced MMP-9 activities were found in treated animals. PDGFR-α inhibition preserved BBB integrity following experimental SAH; however, the protective mechanisms remain to be elucidated. Targeting PDGFR-α signaling might be advantageous to ameliorate early brain injury following SAH.


Subject(s)
Benzamides/therapeutic use , Blood-Brain Barrier/drug effects , Capillary Permeability/drug effects , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Subarachnoid Hemorrhage/pathology , Animals , Blood-Brain Barrier/physiopathology , Brain Edema/drug therapy , Brain Edema/etiology , Capillary Permeability/physiology , Disease Models, Animal , Imatinib Mesylate , Immunoprecipitation , MAP Kinase Kinase 4/metabolism , Male , Neurologic Examination , Proto-Oncogene Proteins pp60(c-src)/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Platelet-Derived Growth Factor alpha/metabolism
18.
Ann Neurol ; 75(2): 209-19, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24273204

ABSTRACT

OBJECTIVE: The NLRP3 (NALP3, cryopyrin) inflammasome, a key component of the innate immune system, facilitates caspase-1 and interleukin (IL)-1ß processing, which amplifies the inflammatory response. Here, we investigated whether NLRP3 knockdown decreases neutrophil infiltration, reduces brain edema, and improves neurological function in an intracerebral hemorrhage (ICH) mouse model. We also determined whether mitochondrial reactive oxygen species (ROS) governed by mitochondrial permeability transition pores (mPTPs) would trigger NLRP3 inflammasome activation following ICH. METHODS: ICH was induced by injecting autologous arterial blood (30µl) into a mouse brain. NLRP3 small interfering RNAs were administered 24 hours before ICH. A mPTP inhibitor (TRO-19622) or a specific mitochondria ROS scavenger (Mito-TEMPO) was coinjected with the blood. In naive animals, rotenone, which is a respiration chain complex I inhibitor, was applied to induce mitochondrial ROS production, and followed by TRO-19622 or Mito-TEMPO treatment. Neurological deficits, brain edema, enzyme-linked immunosorbent assay, Western blot, in vivo chemical cross-linking, ROS assay, and immunofluorescence were evaluated. RESULTS: ICH activated the NLRP3 inflammasome. NLRP3 knockdown reduced brain edema and decreased myeloperoxidase (MPO) levels at 24 hours, and improved neurological functions from 24 to 72 hours following ICH. TRO-19622 or Mito-TEMPO reduced ROS, NLRP3 inflammasome components, and MPO levels following ICH. In naive animals, rotenone administration induced mPTP formation, ROS generation, and NLRP3 inflammasome activation, which were then reduced by TRO-19622 or Mito-TEMPO. INTERPRETATION: The NLRP3 inflammasome amplified the inflammatory response by releasing IL-1ß and promoting neutrophil infiltration following ICH. Mitochondria ROS may be a major trigger of NLRP3 inflammasome activation. The results of our study suggest that the inhibition of the NLRP3 inflammasome may effectively reduce the inflammatory response following ICH.ANN NEUROL 2014;75:209-219.


Subject(s)
Carrier Proteins/metabolism , Cerebral Hemorrhage/complications , Inflammation/etiology , Animals , Antioxidants/therapeutic use , Brain Edema/etiology , Brain Edema/therapy , Carrier Proteins/genetics , Cerebral Hemorrhage/prevention & control , Cholestenones/therapeutic use , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hematoma/drug therapy , Hematoma/etiology , Injections, Intraventricular , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/genetics , Organophosphorus Compounds/therapeutic use , Piperidines/therapeutic use , RNA, Small Interfering/therapeutic use
19.
Stroke ; 44(5): 1410-7, 2013 May.
Article in English | MEDLINE | ID: mdl-23539525

ABSTRACT

BACKGROUND AND PURPOSE: Plasma thrombin concentration is increased after subarachnoid hemorrhage (SAH). However, the role of thrombin receptor (protease-activated receptor-1 [PAR-1]) in endothelial barrier disruption has not been studied. The aims of this study were to investigate the role of PAR-1 in orchestrating vascular permeability and to assess the potential therapeutics of a PAR-1 antagonist, SCH79797, through maintaining vascular integrity. METHODS: SCH79797 was injected intraperitoneally into male Sprauge-Dawley rats undergoing SAH by endovascular perforation. Assessment was conducted at 24 hours after SAH for brain water content, Evans blue content, and neurobehavioral testing. To explore the role of PAR-1 activation and the specific mechanism of SCH79797's effect after SAH, Western blot, immunoprecipitation, and immunofluorescence of hippocampus tissue were performed. A p21-activated kinase-1 (PAK1) inhibitor, IPA-3, was used to explore the underlying protective mechanism of SCH79797. RESULTS: At 24 hours after SAH, animals treated with SCH79797 demonstrated a reduction in brain water content, Evans blue content, and neurobehavioral deficits. SCH79797 also attenuated PAR-1 expression and maintained the level of vascular endothelial-cadherin, an important component of adherens junctions. Downstream to PAR-1, c-Src-dependent activation of p21-activated kinase-1 led to an increased serine/threonine phosphorylation of vascular endothelial-cadherin; immunoprecipitation results revealed an enhanced binding of phosphorylated vascular endothelial-cadherin with endocytosis orchestrator ß-arrestin-2. These pathological states were suppressed after SCH79797 treatment. CONCLUSIONS: PAR-1 activation after SAH increases microvascular permeability, at least, partly through a PAR-1-c-Src-p21-activated kinase-1-vascular endothelial-cadherin phosphorylation pathway. Through suppressing PAR-1 activity, SCH79797 plays a protective role in maintaining microvascular integrity after SAH.


Subject(s)
Blood-Brain Barrier/drug effects , Brain/drug effects , Pyrroles/therapeutic use , Quinazolines/therapeutic use , Receptor, PAR-1/antagonists & inhibitors , Subarachnoid Hemorrhage/drug therapy , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/metabolism , Brain/pathology , Disulfides/pharmacology , Enzyme Inhibitors/pharmacology , Male , Naphthols/pharmacology , Permeability/drug effects , Pyrroles/pharmacology , Quinazolines/pharmacology , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/pathology , p21-Activated Kinases/antagonists & inhibitors
20.
Neurobiol Dis ; 50: 13-20, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23000356

ABSTRACT

Neurosurgical procedures inevitably produce intraoperative hemorrhage. The subsequent entry of blood into the brain parenchyma results in the release of large amounts of thrombin, a known contributor to perihematomal edema formation and apoptosis after brain injury. The present study seeks to test 1) the effect of surgically induced brain injury (SBI) on thrombin activity, expression of thrombin's receptor PAR-1, and PAR-1 mediated apoptosis; 2) the effect of thrombin inhibition by argatroban and PAR-1 inhibition by SCH79797 on the development of secondary brain injury in the SBI model on rats. A total of 88 Sprague-Dawley male rats were randomly divided into sham, vehicle-, argatroban-, or SCH79797-treated groups. SBI involved partial resection of the right frontal lobe under inhalation isoflurane anesthesia. Sham-operated animals received only craniotomy. Thrombin activity, brain water content, and neurological deficits were measured at 24 h following SBI. Involvement of the Ask1/JNK pathway in PAR-1-induced post-SBI apoptosis was characterized by using Ask1 or JNK inhibitors. We observed that SBI increased thrombin activity, yet failed to demonstrate any effect on PAR-1 expression. Argatroban and SCH79797 reduced SBI-induced brain edema and neurological deficits in a dose-dependent manner. SBI-induced apoptosis seemed mediated by the PAR-1/Ask1/JNK pathways. Administration of SCH79797 ameliorated the apoptosis following SBI. Our findings indicate that PAR-1 antagonist protects against secondary brain injury after SBI by decreasing both brain edema and apoptosis by inactivating PAR-1/Ask1/JNK pathway. The anti-apoptotic effect of PAR-1 antagonists may provide a promising path for therapy following SBI.


Subject(s)
Apoptosis/drug effects , Brain Injuries/metabolism , Intraoperative Complications/metabolism , Neurosurgical Procedures/adverse effects , Pyrroles/pharmacology , Quinazolines/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Animals , Apoptosis/physiology , Blotting, Western , Brain/surgery , Brain Edema/etiology , Brain Edema/metabolism , Immunohistochemistry , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , Male , Rats , Rats, Sprague-Dawley , Thrombin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL