Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; 209(Pt A): 153-161, 2022 Jun 01.
Article in English | MEDLINE | ID: mdl-35318077

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune disease with a high incidence. Recent studies have demonstrated that diet can contribute to the development and progression of RA. Indeed, non-starch polysaccharides (NSPs) were known to be related to the improvement of RA. In this study, the collagen-induced rats were administrated with Angelica sinensis polysaccharide (ASP) at 200 mg/kg (L), 400 mg/kg (M), or 800 mg/kg (H). Results showed that ASP could reduce joint swelling and significantly inhibit anti-CII-antibodies and pro-inflammatory factors in RA, H group showed the best treatment among them. Further analysis using 16S rDNA sequencing suggested that ASP could shape the gut microbiota composition. Several key bacteria, including norank_f__norank_o__Clostridia_UCG-014, Lactobacillus, norank_f__Oscillospiraceae, and norank_f__Desulfovibrionaceae, were found to be related to the development of RA. The colonic transcriptome showed that ASP could restore RA-induced intestinal dysfunction, such as tight junction disarrangement, by upregulating Cldn5. The balance between osteoblasts and osteoclasts might be modified by regulating the expression of Slit3 and Rgs18 to alleviate RA, which may be correlated with gut microbiota. Our results suggested that ASP improved RA by regulating gut microbiota and gene expression, revealing a positive relationship between dietary patterns and RA.


Subject(s)
Angelica sinensis , Arthritis, Rheumatoid , Claudin-5 , Gastrointestinal Microbiome , RGS Proteins , Angelica sinensis/chemistry , Angelica sinensis/metabolism , Animals , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Claudin-5/biosynthesis , Claudin-5/genetics , Intestines/metabolism , Intestines/microbiology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Polysaccharides/pharmacology , RGS Proteins/biosynthesis , RGS Proteins/genetics , Rats
2.
Exp Eye Res ; 217: 108977, 2022 04.
Article in English | MEDLINE | ID: mdl-35139333

ABSTRACT

Loss-of-function mutations in the Wnt co-receptor, low-density lipoprotein receptor-related protein 5 (LRP5), result in familial exudative vitreoretinopathy (FEVR), osteoporosis-pseudoglioma syndrome (OPPG), and Norrie disease. CRISPR/Cas9 gene editing was used to produce rat strains deficient in Lrp5. The purpose of this study was to validate this rat model for studies of hypovascular, exudative retinopathies. The retinal vasculature of wildtype and Lrp5 knockout rats was stained with Giffonia simplifolia isolectin B4 and imaged by fluorescence microscopy. Effects on retinal structure were investigated by histology. The integrity of the blood-retina barrier was analyzed by measurement of permeability to Evans blue dye and staining for claudin-5. Retinas were imaged by fundus photography and SD-OCT, and electroretinograms were recorded. Lrp5 gene deletion led to sparse superficial retinal capillaries and loss of the deep and intermediate plexuses. Autofluorescent exudates were observed and are correlated with increased Evans blue permeability and absence of claudin-5 expression in superficial vessels. OCT images show pathology similar to OCT of humans with FEVR, and retinal thickness is reduced by 50% compared to wild-type rats. Histology and OCT reveal that photoreceptor and outer plexiform layers are absent. The retina failed to demonstrate an ERG response. CRISPR/Cas9 gene-editing produced a predictable rat Lrp5 knockout model with extensive defects in the retinal vascular and neural structure and function. This rat model should be useful for studies of exudative retinal vascular diseases involving the Wnt and norrin pathways.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-5 , Retina , Animals , Claudin-5/biosynthesis , Claudin-5/genetics , Evans Blue/pharmacology , Familial Exudative Vitreoretinopathies/genetics , Familial Exudative Vitreoretinopathies/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Mutation , Rats , Retina/metabolism , Structure-Activity Relationship
3.
Can J Cardiol ; 37(10): 1593-1606, 2021 10.
Article in English | MEDLINE | ID: mdl-33838228

ABSTRACT

BACKGROUND: Downregulation of claudin-5 in the heart is associated with the end-stage heart failure. However, the underlying mechanism ofclaudin-5 is unclear. Here we investigated the molecular actions of claudin-5 in perspective of mitochondria in cardiomyocytes to better understand the role of claudin-5 in cardioprotection during ischemia. METHODS: Myocardial ischemia/reperfusion (I/R; 30 min/24 h) and hypoxia/reoxygenation (H/R; 24 h/4 h) were used in this study. Confocal microscopy and transmission electron microscope (TEM) were used to observe mitochondrial morphology. RESULTS: Claudin-5 was detected in murine heart tissue and neonatal rat cardiomyocytes (NRCM). Its protein level was severely decreased after myocardial I/R or H/R. Confocal microscopy showedclaudin-5 presented in the mitochondria of NRCM. H/R-induced claudin-5 downregulation was accompanied by mitochondrial fragmentation. The mitofusin 2 (Mfn2) expressionwas dramatically decreased while the dynamin-related protein (Drp) 1 expression was significantly increased after H/R. The TEM indicatedH/R-induced mitochondrial swelling and fission. Adenoviral claudin-5 overexpression reversed these structural disintegration of mitochondria. The mitochondria-centered intrinsic pathway of apoptosis triggered by H/R and indicated by the cytochrome c and cleaved caspase 3 in the cytoplasm of NRCMs was also reduced by overexpressing claudin-5. Claudin-5 overexpression in mouse heart also significantly decreased cleaved caspase 3 and the infarct size in ischemic heart with improved systolic function. CONCLUSION: We demonstrated for the first time the presence of claudin-5 in the mitochondria in cardiomyocytes and provided the firm evidence for the cardioprotective role of claudin-5 in the preservation of mitochondrial dynamics and cell fate against hypoxia- or ischemia-induced stress.


Subject(s)
Claudin-5/genetics , Hypoxia/prevention & control , Mitochondria, Heart/genetics , Mitochondrial Dynamics/genetics , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , Animals , Animals, Newborn , Apoptosis , Cells, Cultured , Claudin-5/biosynthesis , Dynamins/biosynthesis , Dynamins/genetics , GTP Phosphohydrolases/biosynthesis , GTP Phosphohydrolases/genetics , Hypoxia/genetics , Hypoxia/pathology , Membrane Proteins , Microscopy, Electron, Transmission , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/ultrastructure , Rats , Rats, Sprague-Dawley
4.
Neurogastroenterol Motil ; 33(6): e14084, 2021 06.
Article in English | MEDLINE | ID: mdl-33497497

ABSTRACT

BACKGROUND: Simultaneous evaluation of barrier protein expression in the gut and the brain and their modulation under stress conditions have not been studied before now. As the permeability and function of the gut and blood-brain barrier are different and both express the MRs, we hypothesized that stress of post-weaning social isolation induces changes in tight junction protein expression in the gut which are (1) independent of changes in the brain and (2) are mediated via the mineralocorticoid receptor (MR). METHODS: First, using UPLC-MS/MS we have successfully validated and selected a dose (1.2 mg/rat/day) of the MR antagonist spironolactone to treat female rats exposed to stress of chronic isolation or control conditions from postnatal day 21 for 9 weeks. KEY RESULTS: Isolation stress caused an enhancement of gene expression of occludin and ZO-1 and a decrease in claudin-5 and MR expression in both the small intestine and prefrontal cortex. Isolation stress failed to decrease claudin-5 (small intestine) and MR (prefrontal cortex) gene expression in spironolactone-treated rats. MR blockade resulted in a decrease in claudin-15 expression in the small intestine. Anxiogenic effect of chronic stress, measured in elevated plus-maze test, was partly prevented by spironolactone treatment. CONCLUSIONS & INFERENCES: Claudins, the main regulators of intestinal barrier permeability responded to chronic stress of social isolation and/or simultaneous blockade of MR in female rats by alterations independent of changes in the brain cortex. The results suggest a physiological role of MR in the control of claudin expression in the small intestine, but not in the brain cortex.


Subject(s)
Intestine, Small/metabolism , Prefrontal Cortex/metabolism , Social Isolation , Stress, Psychological/metabolism , Tight Junction Proteins/metabolism , Animals , Anxiety/psychology , Claudin-5/biosynthesis , Claudin-5/genetics , Female , Mineralocorticoid Receptor Antagonists/pharmacology , Occludin/biosynthesis , Occludin/genetics , Rats , Rats, Sprague-Dawley , Spironolactone/pharmacology , Stress, Psychological/psychology , Zonula Occludens-1 Protein/biosynthesis , Zonula Occludens-1 Protein/genetics
5.
Diabetologia ; 64(1): 226-239, 2021 01.
Article in English | MEDLINE | ID: mdl-33106900

ABSTRACT

AIMS/HYPOTHESIS: Microvascular endothelial hyperpermeability, mainly caused by claudin-5 deficiency, is the initial pathological change that occurs in diabetes-associated cardiovascular disease. The ketone body ß-hydroxybutyrate (BHB) exerts unique beneficial effects on the cardiovascular system, but the involvement of BHB in promoting the generation of claudin-5 to attenuate cardiac microvascular hyperpermeability in diabetes is poorly understood. METHODS: The effects of BHB on cardiac microvascular endothelial hyperpermeability and claudin-5 generation were evaluated in rats with streptozotocin-induced diabetes and in high glucose (HG)-stimulated human cardiac microvascular endothelial cells (HCMECs). To explore the underlying mechanisms, we also measured ß-catenin nuclear translocation, binding of ß-catenin, histone deacetylase (HDAC)1, HDAC3 and p300 to the Claudin-5 (also known as CLDN5) promoter, interaction between HDAC3 and ß-catenin, and histone acetylation in the Claudin-5 promoter. RESULTS: We found that 10 weeks of BHB treatment promoted claudin-5 generation and antagonised cardiac microvascular endothelial hyperpermeability in rat models of diabetes. Meanwhile, BHB promoted claudin-5 generation and inhibited paracellular permeability in HG-stimulated HCMECs. Specifically, BHB (2 mmol/l) inhibited HG-induced HDAC3 from binding to the Claudin-5 promoter, although nuclear translocation or promoter binding of ß-catenin did not change with BHB treatment. In addition, BHB prevented the binding and co-localisation of HDAC3 to ß-catenin in HG-stimulated HCMECs. Furthermore, using mass spectrometry, acetylated H3K14 (H3K14ac) in the Claudin-5 promoter following BHB treatment was identified, regardless of whether cells were stimulated by HG or not. Although reduced levels of acetylated H3K9 in the Claudin-5 promoter were found following HG stimulation, increased H3K14ac was specifically associated with BHB treatment. CONCLUSIONS/INTERPRETATION: BHB inhibited HDAC3 and caused acetylation of H3K14 in the Claudin-5 promoter, thereby promoting claudin-5 generation and antagonising diabetes-associated cardiac microvascular hyperpermeability. Graphical abstract.


Subject(s)
3-Hydroxybutyric Acid/pharmacology , Capillary Permeability/drug effects , Claudin-5/biosynthesis , Coronary Vessels/physiopathology , Diabetes Mellitus, Experimental/physiopathology , Histone Deacetylases/drug effects , Animals , Capillary Permeability/physiology , Claudin-5/genetics , Diabetes Complications/prevention & control , Endothelium, Vascular/physiopathology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Male , Microvessels/physiopathology , Promoter Regions, Genetic/physiology , Rats , Rats, Sprague-Dawley , beta Catenin/metabolism
6.
Int Immunol ; 33(3): 171-182, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33038259

ABSTRACT

T-cell development depends on the thymic microenvironment, in which endothelial cells (ECs) play a vital role. Interestingly, vascular permeability of the thymic cortex is lower than in other organs, suggesting the existence of a blood-thymus barrier (BTB). On the other hand, blood-borne molecules and dendritic cells bearing self-antigens are accessible to the medulla, facilitating central tolerance induction, and continuous T-precursor immigration and mature thymocyte egress occur through the vessels at the cortico-medullary junction (CMJ). We found that claudin-5 (Cld5), a membrane protein of tight junctions, was expressed in essentially all ECs of the cortical vasculatures, whereas approximately half of the ECs of the medulla and CMJ lacked Cld5 expression. An intravenously (i.v.) injected biotin tracer hardly penetrated cortical Cld5+ vessels, but it leaked into the medullary parenchyma through Cld5- vessels. Cld5 expression in an EC cell line caused a remarkable increase in trans-endothelial resistance in vitro, and the biotin tracer leaked from the cortical vasculatures in Cldn5-/- mice. Furthermore, i.v.-injected sphingosine-1 phosphate distributed selectively into the medulla through the Cld5- vessels, probably ensuring the egress of CD3high mature thymocytes from Cld5- vessels at the CMJ. These results suggest that distinct Cld5 expression profiles in the cortex and medulla may control the BTB and the T-cell gateway to blood circulation, respectively.


Subject(s)
Capillary Permeability/physiology , Claudin-5/metabolism , T-Lymphocytes/metabolism , Thymus Gland/metabolism , Tight Junctions/physiology , Animals , Cell Differentiation/immunology , Cell Line , Claudin-5/biosynthesis , Endothelial Cells/metabolism , Lysophospholipids/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Sphingosine/analogs & derivatives , Sphingosine/metabolism , T-Lymphocytes/cytology , Thymocytes/metabolism
7.
Sci Signal ; 13(626)2020 04 07.
Article in English | MEDLINE | ID: mdl-32265338

ABSTRACT

The blood-brain barrier (BBB) maintains a stable brain microenvironment. Breakdown of BBB integrity during cerebral ischemia initiates a devastating cascade of events that eventually leads to neuronal loss. MicroRNAs are small noncoding RNAs that suppress protein expression, and we previously showed that the miR-15a/16-1 cluster is involved in the pathogenesis of ischemic brain injury. Here, we demonstrated that when subjected to experimentally induced stroke, mice with an endothelial cell (EC)-selective deletion of miR-15a/16-1 had smaller brain infarcts, reduced BBB leakage, and decreased infiltration of peripheral immune cells. These mice also showed reduced infiltration of proinflammatory M1-type microglia/macrophage in the peri-infarct area without changes in the number of resolving M2-type cells. Stroke decreases claudin-5 abundance, and we found that EC-selective miR-15a/16-1 deletion enhanced claudin-5 mRNA and protein abundance in ischemic mouse brains. In cultured mouse brain microvascular ECs (mBMECs), the miR-15a/16-1 cluster directly bound to the 3' untranslated region (3'UTR) of Claudin-5, and lentivirus-mediated ablation of miR-15a/16-1 diminished oxygen-glucose deprivation (OGD)-induced down-regulation of claudin-5 mRNA and protein abundance and endothelial barrier dysfunction. These findings suggest that genetic deletion of endothelial miR-15a/16-1 suppresses BBB pathologies after ischemic stroke. Elucidating the molecular mechanisms of miR-15a/16-1-mediated BBB dysfunction may enable the discovery of new therapies for ischemic stroke.


Subject(s)
Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Gene Deletion , Ischemic Stroke/metabolism , MicroRNAs/metabolism , Animals , Blood-Brain Barrier/pathology , Claudin-5/biosynthesis , Claudin-5/genetics , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Ischemic Stroke/genetics , Ischemic Stroke/pathology , Ischemic Stroke/prevention & control , Mice , Mice, Knockout , MicroRNAs/genetics
8.
Acta Biochim Biophys Sin (Shanghai) ; 52(4): 363-370, 2020 Apr 20.
Article in English | MEDLINE | ID: mdl-32141492

ABSTRACT

Glycogen synthase kinase-3ß (GSK-3ß), a serine/threonine protein kinase, is widely distributed in mammalian brains. Since GSK-3ß plays a vital role in the development of neurodegenerative disorders, the present study was designed to investigate the role of GSK-3ß in the blood-brain barrier (BBB) permeability in aged mice. Morris water maze test was used to examine mouse cognitive function. BBB permeability was examined by the leakage of fluorescence signals of low-molecular weight dextran. GSK-3ß inhibitor, 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8), was administrated in aged mice and in cultured mouse brain microvascular endothelial cells (bEnd.3). Compared with young mice, aged mice had increased leftover signals of dextran in the hippocampus and a lower score in the maze test, suggesting that aged mice have abnormal leakage of BBB and cognitive dysfunction. The protein expression of Toll-like receptor 4 (TLR4) was increased, whereas the protein expressions of junction proteins (claudin1 and claudin5) were reduced in endothelial cells of BBB in aged mice. Phosphorylated level of serine 9, an inhibitory residue in GSK-3ß protein, was decreased. TDZD-8 treatment downregulated TLR4 protein expression, upregulated claudin1 and claudin5 protein expressions, and significantly improved cognitive function in aged mice. In bEnd.3 cells, TDZD-8 treatment reduced TLR4 expression and increased claudin5 expression in cells stimulated with lipopolysaccharides. In conclusion, the inhibition of GSK-3ß activity downregulates aging-induced TLR4 expression and restores the BBB integrity, resulting in the improvement of cognitive function in aged mice.


Subject(s)
Aging/drug effects , Blood-Brain Barrier/metabolism , Claudin-1/biosynthesis , Claudin-5/biosynthesis , Cognition/drug effects , Endothelial Cells/metabolism , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Thiadiazoles/pharmacology , Up-Regulation/drug effects , Aging/metabolism , Animals , Cerebellum/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Male , Mice
9.
J Neuroimmunol ; 333: 576967, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31151084

ABSTRACT

Chemokine ligand 26 (CCL26) is a member of the eotaxin family. It works by interacting exclusively with chemokine receptor 3 (CCR3) and acts as an eosinophil-selective chemoattractant. There is an emerging role for eotaxins in autoimmune diseases. Studies have reported that chemokine ligand 11 (CCL11) and CCL26 are upregulated in patients with neuromyelitis optica spectrum disorder (NMOSD) during remission, CCL26 levels appear to be decreased in relapsing-remitting multiple sclerosis (RRMS), whereas CCL26 levels are significantly increased in secondary progressive multiple sclerosis (SPMS), indicating that CCL26 participates in the pathogenesis of multiple sclerosis (MS). We investigated the levels of CCL26, CCR3 and claudin-5 (a marker of changes in BBB (blood-brain barrier) permeability) at different stages of experimental autoimmune encephalomyelitis (EAE) to explore the underlying immune mechanisms of EAE. Our results showed that the levels of CCL26 and CCR3 in EAE rats were significantly increased compared with those in the control group. The levels of CCL26 in the serum and in brain tissues as well as the protein expression of CCR3 in brain tissues were positively correlated with the inflammatory scores of brain tissues from EAE rats and were negatively correlated with the protein expression of claudin-5. We concluded that CCL26, which in turn binds to the receptor CCR3, showed pro-inflammatory effects and aggravated tissue damage involving BBB impairment, especially in the acute stage of EAE. Our study uncovers another possible immunopathological mechanism of MS and provides a possible target for immune therapy.


Subject(s)
Chemokine CCL26/physiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Receptors, CCR3/physiology , Animals , Blood-Brain Barrier , Brain/metabolism , Chemokine CCL26/biosynthesis , Chemokine CCL26/genetics , Claudin-5/biosynthesis , Claudin-5/genetics , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Gene Expression Regulation , Inflammation , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Random Allocation , Rats , Rats, Wistar , Receptors, CCR3/biosynthesis , Receptors, CCR3/genetics , Single-Blind Method
10.
PLoS One ; 14(2): e0211818, 2019.
Article in English | MEDLINE | ID: mdl-30742658

ABSTRACT

The present study aimed to estimate the effect of endurance training, two doses of testosterone, and the combination of these stimuli on the level of the endothelial proteins claudin, occludin, JAM-1, VE-cadherin, ZO-1, ZO-2, and P-glycoprotein in rat spinal cords. Adult male Wistar rats were trained using a motor-driven treadmill for 6 weeks (40-60 min, 5 times per week) and/or were treated for 6 weeks with two doses of testosterone (i.m.; 8 mg/kg or 80 mg/kg body weight). Spinal cords were collected 48 hours after the last training cycle and stored at -80°C. The levels of selected proteins in whole tissue lysates of the spinal cord were measured by western blot. Testosterone-treated trained rats had significantly lower claudin levels than vehicle-treated trained rats. High doses of testosterone resulted in a significant decrease in claudin-5 in untrained rats compared to the control group. Both doses of testosterone significantly reduced occludin levels compared to those in vehicle-treated untrained rats. The JAM-1 level in the spinal cords of both trained and untrained animals receiving testosterone was decreased in a dose-dependent manner. The JAM-1 level in the trained group treated with high doses of testosterone was significantly higher than that in the untrained rats treated with 80 mg/kg of testosterone. VE-cadherin levels were decreased in all groups receiving testosterone regardless of endurance training and were also diminished in the vehicle-treated group compared to the control group. Testosterone treatment did not exert a significant effect on ZO-1 protein levels. Testosterone and/or training had no significant effects on ZO-2 protein levels in the rat spinal cords. Endurance training increased P-glycoprotein levels in the rat spinal cords. The results suggest that an excessive supply of testosterone may adversely impact the expression of endothelial proteins in the central nervous system, which, in turn, may affect the blood-brain barrier function.


Subject(s)
Endothelium, Vascular/metabolism , Gene Expression Regulation/drug effects , Physical Conditioning, Animal , Physical Endurance/drug effects , Spinal Cord , Testosterone/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Animals , Antigens, CD/biosynthesis , Cadherins/biosynthesis , Cell Adhesion Molecules/biosynthesis , Claudin-5/biosynthesis , Dose-Response Relationship, Drug , Male , Rats , Rats, Wistar , Spinal Cord/chemistry , Spinal Cord/metabolism , Zonula Occludens-1 Protein/biosynthesis , Zonula Occludens-2 Protein/biosynthesis
11.
J Neurosci ; 39(4): 743-757, 2019 01 23.
Article in English | MEDLINE | ID: mdl-30504279

ABSTRACT

Recent evidence suggests that blood-brain barrier (BBB) recovery and reestablishment of BBB impermeability after stroke is incomplete. This could influence stroke recovery, increase the risk of repeat stroke, and be a solid substrate for developing vascular dementia. Although accumulating evidence has defined morphological alterations and underlying mechanisms of tight junction (TJ) changes during BBB breakdown in acute stroke, very little is known about the type of alterations and mechanisms in BBB "leakage" found subacutely or chronically. The current study examined BBB structural alterations during the "BBB leakage" associated with the chronic phase of stroke in male mice and both genders of humans. We found significant upregulation of claudin-1 mRNA and protein, a nonspecific claudin for blood vessels, and downregulation in claudin-5 expression. Morphological and biochemical as well as fluorescence resonance energy transfer and fluorescence recovery after photobleaching analysis of postischemic brain endothelial cells and cells overexpressing claudin-1 indicated that newly synthesized claudin-1 was present on the cell membrane (∼45%), was incorporated into the TJ complex with established interaction with zonula occludens-1 (ZO-1), and was building homophilic cis- and trans-interactions. The appearance of claudin-1 in the TJ complex reduced claudin-5 strands (homophilic claudin-5 cis- and trans-interactions) and claudin-5/ZO-1 interaction affecting claudin-5 incorporation into the TJ complex. Moreover, claudin-1 induction was associated with an endothelial proinflammatory phenotype. Targeting claudin-1 with a specific C1C2 peptide improved brain endothelial barrier permeability and functional recovery in chronic stroke condition. This study highlights a potential "defect" in postischemic barrier formation that may underlie prolonged vessel leakiness.SIGNIFICANCE STATEMENT Although rarely expressed at the normal blood-brain barrier (BBB), claudin-1 is expressed in pathological conditions. Analyzing poststroke human and mouse blood microvessels we have identified that claudin-1 is highly expressed in leaky brain microvessels. Our results reveal that claudin-1 is incorporated in BBB tight junction complex, impeding BBB recovery and causing BBB leakiness during poststroke recovery. Targeting claudin-1 with a claudin-1 peptide improves brain endothelial barrier permeability and consequently functional neurological recovery after stroke.


Subject(s)
Blood-Brain Barrier/pathology , Claudin-1/genetics , Stroke/genetics , Stroke/pathology , Animals , Brain Ischemia/pathology , Claudin-5/biosynthesis , Claudin-5/genetics , Down-Regulation/genetics , Endothelial Cells/pathology , Female , Humans , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Inflammation/pathology , Male , Mice , Tight Junctions/pathology , Zonula Occludens-1 Protein/biosynthesis , Zonula Occludens-1 Protein/genetics
12.
Br J Neurosurg ; 32(2): 141-148, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29357709

ABSTRACT

PURPOSE: Sodium fluorescein (SF) is an ideal dye for intraoperative guided-resection of high-grade gliomas (HGGs). However, it is not well understood whether the SF-guided technique is suitable for different grades of gliomas, and the correlation between fluorescence and pathology is also not yet clear. MATERIALS AND METHODS: In this study, we investigated 28 patients, including 23 patients with HGG and 5 patients with low-grade glioma (LGG). All patients were treated using the SF-guided technique on a Pentero 900 microscope (Carl Zeiss, Oberkochen, Germany). Claudin-5 immunohistochemical (IHC) staining for the tumours and peritumour tissues was analyzed. RESULTS: Intraoperative yellow fluorescence was noted in all the HGGs but not in the LGGs. Claudin-5 expression in the blood brain barrier endothelial cells was downregulated and disconnected in the HGGs (p < 0.05), but had no difference or slightly decreased in the LGGs (p > 0.05). CONCLUSIONS: The SF-guided technique is suitable for HGG surgery but not for LGG surgery. Downregulation of claudin-5 expression may contribute to the presence of yellow fluorescence in the glioma in SF-guided surgery.


Subject(s)
Blood-Brain Barrier/injuries , Brain Neoplasms/surgery , Glioma/surgery , Neurosurgical Procedures/methods , Surgery, Computer-Assisted/methods , Adult , Aged , Brain Neoplasms/diagnostic imaging , Claudin-5/biosynthesis , Contrast Media , Down-Regulation , Female , Fluorescein , Fluorescence , Glioma/diagnostic imaging , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Male , Middle Aged , Neoplasm Grading , Treatment Outcome
13.
Nat Neurosci ; 20(12): 1752-1760, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29184215

ABSTRACT

Studies suggest that heightened peripheral inflammation contributes to the pathogenesis of major depressive disorder. We investigated the effect of chronic social defeat stress, a mouse model of depression, on blood-brain barrier (BBB) permeability and infiltration of peripheral immune signals. We found reduced expression of the endothelial cell tight junction protein claudin-5 (Cldn5) and abnormal blood vessel morphology in nucleus accumbens (NAc) of stress-susceptible but not resilient mice. CLDN5 expression was also decreased in NAc of depressed patients. Cldn5 downregulation was sufficient to induce depression-like behaviors following subthreshold social stress whereas chronic antidepressant treatment rescued Cldn5 loss and promoted resilience. Reduced BBB integrity in NAc of stress-susceptible or mice injected with adeno-associated virus expressing shRNA against Cldn5 caused infiltration of the peripheral cytokine interleukin-6 (IL-6) into brain parenchyma and subsequent expression of depression-like behaviors. These findings suggest that chronic social stress alters BBB integrity through loss of tight junction protein Cldn5, promoting peripheral IL-6 passage across the BBB and depression.


Subject(s)
Depression/pathology , Depression/psychology , Social Environment , Stress, Psychological/pathology , Stress, Psychological/psychology , Adrenergic Uptake Inhibitors/pharmacology , Animals , Anxiety/psychology , Behavior, Animal , Blood-Brain Barrier/pathology , Claudin-5/biosynthesis , Claudin-5/genetics , Feeding Behavior , Food Preferences , Imipramine/pharmacology , Interleukin-6/biosynthesis , Male , Mice , Mice, Inbred C57BL , Nucleus Accumbens/pathology , Swimming/psychology , Tight Junction Proteins/metabolism
14.
Cell Physiol Biochem ; 41(5): 1924-1934, 2017.
Article in English | MEDLINE | ID: mdl-28391269

ABSTRACT

BACKGROUND/AIMS: Although proinflammatory cytokine-induced disruption of intestinal epithelial barrier integrity is associated with intestinal inflammatory disease, effective treatment for barrier dysfunction is lacking. Previously, we demonstrated that rebeccamycin alleviates epithelial barrier dysfunction induced by inflammatory cytokines in Caco-2 cell monolayers; however, the underlying mechanism remained unclear. Here, we investigated the mechanism by which rebeccamycin protects the epithelial barrier function of Caco-2 cells exposed to TNF-α. METHODS: To confirm the epithelial barrier function of Caco-2 cell monolayers, transepithelial electrical resistance (TER) and paracellular permeability were measured. Production levels and localization of tight junction (TJ) proteins were analyzed by immunoblot and immunofluorescence, respectively. Phosphorylated myosin light chain (pMLC) and MLC kinase (MLCK) mRNA expression levels were determined by immunoblot and quantitative RT-PCR, respectively. RESULTS: Rebeccamycin attenuated the TNF-α-induced reduction in TER and increase in paracellular permeability. Rebeccamycin increased claudin-5 expression, but not claudin-1, -2, -4, occludin or ZO-1 expression, and prevented the TNF-α-induced changes in ZO-1 and occludin localization. Rebeccamycin suppressed the TNF-α-induced increase in MLCK mRNA expression, thus suppressing MLC phosphorylation. The rebeccamycin-mediated reduction in MLCK production and protection of epithelial barrier function were alleviated by Chk1 inhibition. CONCLUSION: Rebeccamycin attenuates TNF-α-induced disruption of intestinal epithelial barrier integrity by inducing claudin-5 expression and suppressing MLCK production via Chk1 activation.


Subject(s)
Carbazoles/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Intestinal Mucosa/enzymology , Myosin-Light-Chain Kinase/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , Caco-2 Cells , Checkpoint Kinase 1/metabolism , Claudin-5/biosynthesis , Enzyme Activation/drug effects , Humans , Myosin Light Chains/metabolism , Phosphorylation/drug effects , Tight Junctions/enzymology
15.
Biochim Biophys Acta Mol Basis Dis ; 1863(9): 2240-2254, 2017 09.
Article in English | MEDLINE | ID: mdl-28185956

ABSTRACT

The blood-tumor barrier (BTB) constitutes an efficient organization of tight junctions that limits the delivery of chemotherapeutic drugs to brain tumor tissues and impacts the treatment of glioma. Long non-coding RNAs (lncRNAs) are non-protein coding RNAs regulating gene expression, some lncRNAs play a crucial role in BTB permeability. However, the function of lncRNAs in BTB permeability is still largely unclear. Here, we have identified lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1), was remarkably up-regulated in glioma endothelial cells (GECs) obtained from an in vitro BTB model. Knockdown of NEAT1 impaired the integrity and increased the permeability of the BTB, accompanied by downregulation of expression of the tight junction proteins ZO-1, occludin and claudin-5 in GECs. Both bioinformatics data and results of luciferase reporter assays demonstrated that NEAT1 influenced BTB permeability by binding to miR-181d-5p. Knockdown of NEAT1 also down-regulated the expression of sex determining region Y-box protein 5 (SOX5), which was defined as a direct and functional downstream target of miR-181d-5p. SOX5 interacts with the promoter region of ZO-1, occludin and claudin-5 in GECs. In conclusion, knockdown of NEAT1 increased BTB permeability by binding to miR-181d-5p and then reducing tight junction protein expression by targeting SOX5. These results suggest an important role for NEAT1 in regulating BTB permeability and provide an additional strategy for treating glioma.


Subject(s)
Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Glioma/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Tight Junction Proteins/biosynthesis , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Claudin-5/biosynthesis , Claudin-5/genetics , Gene Knockdown Techniques , Glioma/genetics , Glioma/pathology , HEK293 Cells , Humans , MicroRNAs/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Occludin/biosynthesis , Occludin/genetics , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , SOXD Transcription Factors/genetics , SOXD Transcription Factors/metabolism , Zonula Occludens-1 Protein/biosynthesis , Zonula Occludens-1 Protein/genetics
16.
J Cereb Blood Flow Metab ; 37(1): 85-96, 2017 01.
Article in English | MEDLINE | ID: mdl-26661237

ABSTRACT

Stroke is a disease in dire need of better therapies. We have previously shown that a fragment of the extracellular matrix proteoglycan, perlecan, has beneficial effects following cerebral ischemia via the α5ß1 integrin receptor. We now report that endothelial cell selective α5 integrin deficient mice (α5 KO) are profoundly resistant to ischemic infarct after transient middle cerebral artery occlusion. Specifically, α5 KOs had little to no infarct 2-3 days post-stroke, whereas controls had an increase in mean infarct volume over the same time period as expected. Functional outcome is also improved in the α5 KOs compared with controls. Importantly, no differences in cerebrovascular anatomy or collateral blood flow were noted that could account for this difference in ischemic injury. Rather, we demonstrate that α5 KOs have increased blood-brain barrier integrity (increased expression of claudin-5, and absent brain parenchymal IgG extravasation) after stroke compared with controls, which could explain their resistance to ischemic injury. Additionally, inhibition of α5 integrin in vitro leads to decreased permeability of brain endothelial cells following oxygen-glucose deprivation. Together, these findings indicate endothelial cell α5 integrin plays an important role in stroke outcome and blood-brain barrier integrity, suggesting that α5 integrin could be a novel therapeutic target for stroke.


Subject(s)
Brain Ischemia/pathology , Disease Susceptibility/chemically induced , Integrin alpha5/physiology , Stroke/etiology , Animals , Blood-Brain Barrier/metabolism , Brain Infarction/pathology , Claudin-5/biosynthesis , Disease Models, Animal , Endothelial Cells/metabolism , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Integrin alpha5/genetics , Mice , Mice, Knockout , Stroke/pathology
17.
Pathol Res Pract ; 212(11): 959-964, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27665022

ABSTRACT

Nitric oxide (NO) overproduction has been demonstrated from different NO-synthase overexpression or hyperactivity after brain ischemia. Here, we examined the effects of inhibition of NO overproduction on brain infarction, cerebrovascular damage and expression of claudin-5 and zonula occludens-1 (ZO-1) in striatum of ischemic brain. The experiment was performed in three groups of rats; sham, control ischemia and ischemic treatment. Brain ischemia was induced by 60min of middle cerebral artery occlusion (MCAO) followed by 24h of reperfusion. Treated rats received L-NAME 30min before induction of ischemia (1mg/kg, i.p.). Infarct volume and histopathological changes of ischemic striatum were assessed by TTC and LFB staining methods, respectively. Ultimately, quantitative RT-PCR was used for assessment of claudins-5 and ZO-1 expression. MCAO in the control group induced infarction (135±25mm3) at large areas of striatum in accompany with neuronal damages, whereas L-NAME significantly reduced infarction (87±16mm3) and neuronal injuries. The mRNA of ZO-1 and claudin-5 decreased in ischemic striatum, whereas inhibition of NO overproduction by L-NAME attenuated this reduction for these genes. Our findings indicated that NO overproduction after brain ischemia plays a crucial role in neuronal damage especially at striatal regions. Hence, inhibition of excessive NO production may save striatal cerebrovascular integrity of ischemic brain.


Subject(s)
Brain Infarction/pathology , Claudin-5/biosynthesis , Nitric Oxide/biosynthesis , Zonula Occludens-1 Protein/biosynthesis , Animals , Brain Infarction/metabolism , Capillary Permeability/physiology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley
18.
Exp Eye Res ; 151: 75-81, 2016 10.
Article in English | MEDLINE | ID: mdl-27523466

ABSTRACT

Diabetic macular oedema (DMO), a leading cause of preventable visual loss in the working population, is caused by an increase in microvascular endothelial cell permeability, and its prevalence is on the increase in parallel with the rising worldwide prevalence of diabetes. It is known that retinal vascular leakage in DMO is contributed to by VEGF upregulation as well as non-VEGF dependent inflammatory pathways, and the potential use of anti-inflammatory agents such as the glucocorticoids, including dexamethasone are being extensively studied. However, the mechanisms of action of dexamethasone in DMO reduction are not fully understood. Using human primary retinal endothelial cells (REC) the in vitro effect of dexamethasone in modulating the proliferation, permeability and gene expression of key tight and adheren junction components, and the expression of angiopoietins (Ang) 1 and 2 in high (25 mM) glucose conditions were investigated. High glucose decreased REC proliferation, an effect that was reversed by dexamethasone. High glucose conditions significantly increased REC permeability and decreased claudin-5, occludin and JAM-A gene expression; dexamethasone was effective in partially reversing these changes, restoring EC permeability to the normal or near normal state. High glucose levels resulted in reduction of Ang1 secretion, although Ang2 levels were consistently high. DEX increased Ang1 and decreased Ang2, indicating that the balance of Ang1/Ang2 may be important in determining functional changes in REC under high glucose conditions.


Subject(s)
Cell Membrane Permeability/drug effects , Dexamethasone/pharmacokinetics , Diabetic Retinopathy/drug therapy , Endothelial Cells/metabolism , Glucose/pharmacokinetics , Macular Edema/drug therapy , Retina/metabolism , Aged , Aged, 80 and over , Animals , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cell Proliferation , Cells, Cultured , Claudin-5/biosynthesis , Claudin-5/genetics , Dexamethasone/administration & dosage , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation/drug effects , Glucocorticoids/administration & dosage , Glucocorticoids/pharmacokinetics , Glucose/administration & dosage , Humans , Macular Edema/metabolism , Macular Edema/pathology , Male , Middle Aged , RNA/genetics , Real-Time Polymerase Chain Reaction , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Retina/drug effects , Retina/pathology , Ribonuclease, Pancreatic/metabolism , Sweetening Agents/administration & dosage , Sweetening Agents/pharmacokinetics
19.
Neurotoxicology ; 52: 64-71, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26582458

ABSTRACT

Prenatal exposure to environmental chemicals such as dioxins is known to have adverse effects on the developing central nervous system (CNS) in mammals. Because the fetal blood-brain barrier (BBB) is immature, dioxins are thought to exert their toxic effects on the CNS by crossing the BBB and acting on neural cells directly. However, little is known whether dioxins alter the BBB. In this study, to investigate the effects of dioxins on BBB function, we exposed an in vitro BBB system comprising rat endothelial cells, astrocytes, and pericytes to the toxicant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) either before or after BBB formation. We assessed BBB permeability and the function of tight junctions by measuring transendothelial electric resistance (TEER) values following exposure. Subsequently, total RNA and proteins were obtained from the cells for analysis. TEER values following TCDD exposure before but not after BBB formation were lower than those of the control group. We also observed that the expression of the tight junction proteins ZO-1 and claudin-5 was suppressed following TCDD exposure. To examine the cause of this reduction in protein levels, we performed a real-time quantitative polymerase chain reaction assay and observed low expression of the glial cell line-derived neurotrophic factor (GDNF) mRNA in the exposed groups. Moreover, to rescue the effects of TCDD, we applied extrinsic GDNF with TCDD. The several disruptions caused by TCDD were rescued by the GDNF addition. Our findings suggest that exposure to TCDD during BBB formation disrupts and impairs BBB function in part by the suppression of GDNF action, which may contribute to the adverse effects of TCDD on the fetal CNS.


Subject(s)
Blood-Brain Barrier/drug effects , Polychlorinated Dibenzodioxins/toxicity , Animals , Astrocytes/metabolism , Blood-Brain Barrier/growth & development , Blood-Brain Barrier/pathology , Claudin-5/biosynthesis , Coculture Techniques , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1B1/genetics , Electric Impedance , Endothelial Cells/metabolism , Glial Cell Line-Derived Neurotrophic Factor/genetics , In Vitro Techniques , Models, Biological , Pericytes/metabolism , Polychlorinated Dibenzodioxins/antagonists & inhibitors , RNA, Messenger/analysis , Rats , Zonula Occludens-1 Protein/biosynthesis
20.
Biochim Biophys Acta ; 1859(2): 324-38, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26619802

ABSTRACT

The blood-tumor barrier (BTB) forms a major obstacle in brain tumor therapy by preventing the delivery of sufficient quantities of therapeutic drugs. Long non-coding RNAs (lncRNAs) play important roles in both normal development and diseases including cancer. Here, we elucidated the expression of lncRNA metastasis associated lung adenocarcinoma transcript 1 (MALAT1) and defined its functional role in the regulation of BTB function as well as its possible molecular mechanisms. Our results proved that MALAT1 expression was up-regulated in brain microvessels of human glioma and glioma endothelial cells (GECs) which were obtained by co-culturing endothelial cells with glioma cells. Functionally, knockdown of MALAT1 resulted in an impairment and increased the permeability of BTB as well as decreased the expression of ZO-1, occludin and claudin-5 in GECs. Further, there was reciprocal repression between MALAT1 and miR-140, and miR-140 mediated the effects that MALAT1 knockdown exerted. Mechanistic investigations defined that nuclear factor YA (NFYA), a CCAAT box-binding transcription factor, was a direct and functional downstream target of miR-140, which was involved in the MALAT1 knockdown induced regulation of BTB function. Furthermore, NFYA could up-regulate the promoter activities and bind to the promoters of ZO-1, occludin and claudin-5 in GECs. Taken together, we have demonstrated the fact that knockdown of MALAT1 resulted in the increased permeability of BTB, which might contribute to establishing potential therapeutic strategies for human gliomas.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , MicroRNAs/biosynthesis , RNA, Long Noncoding/genetics , Blood-Brain Barrier/pathology , Brain Neoplasms/pathology , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , Capillary Permeability/genetics , Cell Line, Tumor , Claudin-5/biosynthesis , Claudin-5/genetics , Endothelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioma/pathology , Humans , MicroRNAs/genetics , Occludin/genetics , Promoter Regions, Genetic , Zonula Occludens-1 Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...