Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
Microvasc Res ; 135: 104144, 2021 05.
Article in English | MEDLINE | ID: mdl-33515567

ABSTRACT

Ischemic stroke is a leading cause of morbidity and mortality worldwide, with oxidative stress playing a key role in the injury mechanism of thrombolytic therapy. There is increasing evidence that oxidative stress damages endothelial cells (ECs), degrades tight junction proteins (TJs), and contributes to increased blood-brain barrier (BBB) permeability. It has been demonstrated that the breakdown of BBB could increase the risk of intracerebral hemorrhagic transformation in ischemic stroke. And an episode of cerebral ischemia/reperfusion (I/R) also initiates oxidative stress-mediated inflammatory processes in ECs, which further promotes BBB disruption and the progression of brain injury. Previous studies have revealed that antioxidants could inhibit ROS generation and attenuate BBB disruption after cerebral I/R. Peroxiredoxin 4 (Prx4) is a member of the antioxidant enzymes family (Prx1-6) and has been characterized to be an efficient H2O2 scavenger. It should be noted that Prx4 may be directly involved in the protection of ECs from the effects of ROS and function in ECs as a membrane-associated peroxidase. This paper reviewed the implication of Prx4 on vascular integrity and neuroinflammation following a cerebral I/R injury.


Subject(s)
Blood-Brain Barrier/enzymology , Capillary Permeability , Endothelial Cells/enzymology , Inflammation Mediators/metabolism , Ischemic Stroke/enzymology , Neuroimmunomodulation , Peroxiredoxins/metabolism , Reperfusion Injury/enzymology , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/pathology , Endothelial Cells/immunology , Endothelial Cells/pathology , Humans , Ischemic Stroke/immunology , Ischemic Stroke/pathology , Oxidative Stress , Reactive Oxygen Species/metabolism , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Signal Transduction , Tight Junctions/enzymology , Tight Junctions/immunology , Tight Junctions/pathology
2.
Biomed Pharmacother ; 129: 110415, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32603892

ABSTRACT

Tight junctions play an important role in maintaining barrier integrity of intestinal epithelia. Activation of AMP-activated protein kinase (AMPK) promotes tight junction assembly in intestinal epithelial cells (IEC). Fructo-oligosaccharides (FOS), well-known prebiotics, have previously been shown to alleviate inflammation-associated intestinal epithelial disruption although the mechanisms were unclear. This study aimed to investigate any effect of FOS on AMPK activity and tight junction assembly under non-inflammatory and inflammatory conditions using T84 cells as an IEC model. As analyzed by western blot, FOS induced AMPK activation through a calcium sensing receptor (CaSR)-phospholipase C (PLC)- Ca2+/calmodulin-dependent protein kinase kinase-ß (CaMKKß) pathway. Calcium switch assays and immunofluorescence staining of zonula occludens-1 (ZO-1) revealed that FOS induced tight junction assembly via an CaMKKß-AMPK-dependent mechanism in IEC. Interestingly, FOS reversed the suppressive effect of lipopolysaccharide (LPS) on AMPK activity and tight junction assembly via a CaMKKß pathway. Taken together, these findings uncover a prebiotic-independent effect of FOS in promoting intestinal epithelial tight junction assembly through AMPK activation, which may have implications for the treatment of diseases whose pathogenesis involves impaired intestinal barrier function.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Intestinal Mucosa/drug effects , Oligosaccharides/pharmacology , Prebiotics , Tight Junctions/drug effects , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cell Line , Claudin-1/metabolism , Humans , Intestinal Mucosa/enzymology , Lipopolysaccharides/pharmacology , Occludin/metabolism , Phosphorylation , Receptors, Calcium-Sensing/metabolism , Tight Junctions/enzymology , Type C Phospholipases/metabolism , Zonula Occludens-1 Protein/metabolism
3.
Am J Physiol Cell Physiol ; 318(3): C486-C501, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31913699

ABSTRACT

AMP-activated protein kinase (AMPK) activation promotes early stages of epithelial junction assembly. AMPK activation in MDCK renal epithelial cells facilitates localization of the junction-associated proteins aPKCζ and Par3 to the plasma membrane and promotes conversion of Cdc42, a key regulator of epithelial polarization and junction assembly, to its active GTP bound state. Furthermore, Par3 is an important regulator of AMPK-mediated aPKCζ localization. Both aPKCζ and Par3 serve as intermediates in AMPK-mediated junction assembly, with inhibition of aPKCζ activity or Par3 knockdown disrupting AMPK's ability to facilitate zonula occludens (ZO-1) localization. AMPK phosphorylates the adherens junction protein afadin and regulates its interaction with the tight-junction protein zonula occludens-1. Afadin is phosphorylated at two critical sites, S228 (residing within an aPKCζ consensus site) and S1102 (residing within an AMPK consensus site), that are differentially regulated during junction assembly and that exert different effects on the process. Expression of phospho-defective mutants (S228A and S1102A) perturbed ZO-1 localization to the plasma membrane during AMPK-induced junction assembly. Expression of S228A increased the ZO-1/afadin interaction, while S1102A reduced this interaction during extracellular calcium-induced junction assembly. Inhibition of aPKCζ activity also increased the ZO-1/afadin interaction. Taken together, these data suggest that aPKCζ phosphorylation of afadin terminates the ZO-1/afadin interaction and thus permits the later stages of junction assembly.


Subject(s)
AMP-Activated Protein Kinases/physiology , Cell Membrane/enzymology , Tight Junctions/enzymology , Animals , Cell Membrane/chemistry , Dogs , Madin Darby Canine Kidney Cells , Mice , Phosphorylation/physiology , Protein Kinase C/metabolism , Tight Junctions/chemistry , Zonula Occludens-1 Protein/metabolism
4.
Cardiovasc Pathol ; 41: 11-17, 2019.
Article in English | MEDLINE | ID: mdl-31004933

ABSTRACT

Atherosclerosis is a chronic inflammatory disease with lipid accumulation. Apolipoprotein C3 (APOC3), which is an important regulator of human lipid metabolism, is associated with multiple vascular mechanisms in atherosclerosis and proinflammatory responses. We have previously reported that the expression of inflammatory cytokine TNF-α is elevated in human endothelial cells (HUVECs) after APOC3 treatment. This study investigates the APOC3 signaling pathway involved in TNF-α-mediated expression of JAM-1 in HUVECs. Cultured HUVECs were exposed to APOC3 (50 µg/ml) for 16 h. Mechanistic studies were carried out by silencing TNF-α gene with lentiviral TNF-α-shRNA. Our study was based on the eight signaling pathway inhibitors to block the effect of APOC3 in HUVECs. The expression of JAM-1 was determined by qRT-PCR, Western blotting, and flow cytometry. IKK2 degradation and NF-κB p65 phosphorylation were determined by Western blotting. Our results showed that APOC3 significantly promoted the TNF-α-induced expression of JAM-1 in HUVECs. Inhibiting APOC3 reversed the TNF-α-induced overexpression of JAM-1. Moreover, APOC3 induced the expression of NF-κB p65 and degraded IκB. In conclusion, APOC3 promoted the expression of JAM-1 via the NF-κB, IKK2, and PI3K signaling pathway.


Subject(s)
Apolipoprotein C-III/pharmacology , Cell Adhesion Molecules/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , I-kappa B Kinase/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Receptors, Cell Surface/metabolism , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cell Adhesion Molecules/genetics , Cells, Cultured , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Phosphorylation , Proteolysis , RNA Interference , Receptors, Cell Surface/genetics , Signal Transduction/drug effects , Tight Junctions/drug effects , Tight Junctions/enzymology , Tumor Necrosis Factor-alpha/genetics
5.
Gastroenterology ; 155(4): 1250-1263.e5, 2018 10.
Article in English | MEDLINE | ID: mdl-29928898

ABSTRACT

BACKGROUND & AIMS: Pancreatitis after endoscopic retrograde cholangiopancreatography (PEP) is thought to be provoked by pancreatic ductal hypertension, via unknown mechanisms. We investigated the effects of hydrostatic pressures on the development of pancreatitis in mice. METHODS: We performed studies with Swiss Webster mice, B6129 mice (controls), and B6129 mice with disruption of the protein phosphatase 3, catalytic subunit, ßisoform gene (Cnab-/- mice). Acute pancreatitis was induced in mice by retrograde biliopancreatic ductal or intraductal infusion of saline with a constant hydrostatic pressure while the proximal common bile duct was clamped -these mice were used as a model of PEP. Some mice were given pancreatic infusions of adeno-associated virus 6-nuclear factor of activated T-cells-luciferase to monitor calcineurin activity or the calcineurin inhibitor FK506. Blood samples and pancreas were collected at 6 and 24 hours and analyzed by enzyme-linked immunosorbent assay, histology, immunohistochemistry, or fluorescence microscopy. Ca2+ signaling and mitochondrial permeability were measured in pancreatic acinar cells isolated 15 minutes after PEP induction. Ca2+-activated phosphatase calcineurin within the pancreas was tracked in vivo over 24 hours. RESULTS: Intraductal pressures of up to 130 mm Hg were observed in the previously reported model of PEP; we found that application of hydrostatic pressures of 100 and 150 mm Hg for 10 minutes consistently induced pancreatitis. Pancreatic tissues had markers of inflammation (increased levels of interleukin [IL] 6, IL1B, and tumor necrosis factor), activation of signal transducer and activator of transcription 3, increased serum amylase and IL6, and loss of tight junction integrity. Transiently high pressures dysregulated Ca2+ processing (reduced Ca2+ oscillations and an increased peak plateau Ca2+ signal) and reduced the mitochondrial membrane potential. We observed activation of pancreatic calcineurin in the pancreas in mice. Cnab-/- mice, which lack the catalytic subunit of calcineurin, and mice given FK506 did not develop pressure-induced pancreatic inflammation, edema, or loss of tight junction integrity. CONCLUSIONS: Transient high ductal pressure produces pancreatic inflammation and loss of tight junction integrity in a mouse model of PEP. These processes require calcineurin signaling. Calcineurin inhibitors might be used to prevent acute pancreatitis that results from obstruction.


Subject(s)
Ampulla of Vater/enzymology , Calcineurin/metabolism , Calcium Signaling , Mechanotransduction, Cellular , Pancreatitis/enzymology , Tight Junctions/enzymology , Ampulla of Vater/drug effects , Ampulla of Vater/pathology , Amylases/blood , Animals , Calcineurin/deficiency , Calcineurin/genetics , Calcineurin Inhibitors/pharmacology , Calcium Signaling/drug effects , Cholangiopancreatography, Endoscopic Retrograde , Disease Models, Animal , Female , Hydrostatic Pressure , Interleukin-1beta/metabolism , Interleukin-6/blood , Male , Mechanotransduction, Cellular/drug effects , Membrane Potential, Mitochondrial , Mice, Knockout , Mitochondria/metabolism , Pancreatitis/etiology , Pancreatitis/pathology , Pancreatitis/prevention & control , STAT3 Transcription Factor/metabolism , Tacrolimus/pharmacology , Tight Junctions/drug effects , Tight Junctions/pathology , Time Factors , Tumor Necrosis Factor-alpha/metabolism
6.
Am J Physiol Heart Circ Physiol ; 314(4): H693-H703, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29351469

ABSTRACT

An intact blood-brain barrier (BBB) limits entry of proinflammatory and neurotoxic blood-derived factors into the brain parenchyma. The BBB is damaged in Alzheimer's disease (AD), which contributes significantly to the progression of AD pathologies and cognitive decline. However, the mechanisms underlying BBB breakdown in AD remain elusive, and no interventions are available for treatment or prevention. We and others recently established that inhibition of the mammalian/mechanistic target of rapamycin (mTOR) pathway with rapamycin yields significant neuroprotective effects, improving cerebrovascular and cognitive function in mouse models of AD. To test whether mTOR inhibition protects the BBB in neurological diseases of aging, we treated hAPP(J20) mice modeling AD and low-density lipoprotein receptor-null (LDLR-/-) mice modeling vascular cognitive impairment with rapamycin. We found that inhibition of mTOR abrogates BBB breakdown in hAPP(J20) and LDLR-/- mice. Experiments using an in vitro BBB model indicated that mTOR attenuation preserves BBB integrity through upregulation of specific tight junction proteins and downregulation of matrix metalloproteinase-9 activity. Together, our data establish mTOR activity as a critical mediator of BBB breakdown in AD and, potentially, vascular cognitive impairment and suggest that rapamycin and/or rapalogs could be used for the restoration of BBB integrity. NEW & NOTEWORTHY This report establishes mammalian/mechanistic target of rapamycin as a critical mediator of blood-brain barrier breakdown in models of Alzheimer's disease and vascular cognitive impairment and suggests that drugs targeting the target of rapamycin pathway could be used for the restoration of blood-brain barrier integrity in disease states.


Subject(s)
Alzheimer Disease/drug therapy , Behavior, Animal , Blood-Brain Barrier/drug effects , Cognition , Dementia, Vascular/drug therapy , Protein Kinase Inhibitors/pharmacology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Blood-Brain Barrier/enzymology , Blood-Brain Barrier/pathology , Cell Line , Dementia, Vascular/enzymology , Dementia, Vascular/pathology , Dementia, Vascular/psychology , Disease Models, Animal , Female , Male , Matrix Metalloproteinase 9/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/deficiency , Receptors, LDL/genetics , TOR Serine-Threonine Kinases/metabolism , Tight Junction Proteins/metabolism , Tight Junctions/drug effects , Tight Junctions/enzymology , Tight Junctions/pathology
7.
Mol Neurobiol ; 55(3): 2577-2590, 2018 03.
Article in English | MEDLINE | ID: mdl-28421532

ABSTRACT

After spinal cord injury (SCI), tight junction (TJ) protein degradation increases permeability and disrupts the blood-spinal cord barrier (BSCB). The BSCB is primarily formed of endothelial cell, which forms a specialized tight seal due to the presence of TJs. BSCB disruption after SCI allows neutrophil infiltration. Matrix metalloproteinase (MMP)-8 is believed to be mainly expressed by neutrophils and is quickly released upon neutrophil activation. Here, we determined whether MMP-8 is involved in the TJ protein degradation in endothelial cells and also determined its role in the neuroinflammation after SCI. MMP-8 recombinant protein treatment increases the TNF-α expression and decreased the TJ (occludin and zonula occludens-1) protein expression in the endothelial cells. Likewise, specific MMP-8 inhibitor (MMP-8I) significantly prevented the TNF-α-induced decrease in the expression of TJ protein in endothelial cells. Furthermore, MMP-8 expression was significantly increased 1 and 3 days after moderate compression (35 g for 5 min at T10 level) SCI, whereas TJ protein levels decreased as determined qRT-PCR, western blotting, and immunohistochemistry. MMP-8 was inhibited directly using a MMP-8I (5 mg/kg) and indirectly by reducing neutrophil infiltration with sivelestat sodium (50 mg/kg) or using the antioxidant N-acetyl-L-cysteine (100 mg/kg). The MMP-8I significantly decreased TNF-α expression, IL-6, and iNOS expression and increased TJ protein expression after SCI. In addition, MMP-8I significantly lessens the amount of Evans blue dye extravasation observed after injury. Thus, our result suggests that MMP-8 plays an imperative role in inflammation and degradation of TJ proteins. Increased MMP-8 expression was associated with the early inflammatory phase of SCI. Inhibiting MMP-8 significantly attenuated SCI-induced inflammation, BSCB breakdown, and cell injury.


Subject(s)
Blood-Brain Barrier/enzymology , Disease Models, Animal , Matrix Metalloproteinase 8/metabolism , Matrix Metalloproteinase Inhibitors/therapeutic use , Spinal Cord Injuries/enzymology , Spinal Cord/enzymology , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Dose-Response Relationship, Drug , Female , Inflammation/drug therapy , Inflammation/enzymology , Inflammation/pathology , Matrix Metalloproteinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/prevention & control , Tight Junctions/drug effects , Tight Junctions/enzymology , Tight Junctions/pathology
8.
J Cell Physiol ; 233(1): 186-200, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28639275

ABSTRACT

The MEKK3/MEK5/ERK5 signaling axis is required for cardiovascular development in vivo. We analyzed the physiological role of ERK5 in cardiac endothelial cells and the consequence of activation of this kinase by the statin class of HMG Co-A reductase inhibitor drugs. We utilized human cardiac microvascular endothelial cells (HCMECs) and altered ERK5 expression using siRNA mediated gene silencing or overexpression of constitutively active MEK5 and ERK5 to reveal a role for ERK5 in regulating endothelial tight junction formation and cell permeability. Statin treatment of HCMECs stimulated activation of ERK5 and translocation to the plasma membrane resulting in co-localization with the tight junction protein ZO-1 and a concomitant reduction in endothelial cell permeability. Statin mediated activation of ERK5 was a consequence of reduced isoprenoid synthesis following HMG Co-A reductase inhibition. Statin pretreatment could overcome the effect of doxorubicin in reducing endothelial tight junction formation and prevent increased permeability. Our data provide the first evidence for the role of ERK5 in regulating endothelial tight junction formation and endothelial cell permeability. Statin mediated ERK5 activation and the resulting decrease in cardiac endothelial cell permeability may contribute to the cardioprotective effects of statins in reducing doxorubicin-induced cardiotoxicity.


Subject(s)
Capillary Permeability/drug effects , Coronary Vessels/drug effects , Endothelial Cells/drug effects , Heart Diseases/prevention & control , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mitogen-Activated Protein Kinase 7/metabolism , Tight Junctions/drug effects , Antibiotics, Antineoplastic/toxicity , Cardiotoxicity , Cells, Cultured , Coronary Vessels/enzymology , Cytoprotection , Dose-Response Relationship, Drug , Doxorubicin/toxicity , Endothelial Cells/enzymology , Enzyme Activation , Heart Diseases/chemically induced , Heart Diseases/enzymology , Heart Diseases/genetics , Humans , Mitogen-Activated Protein Kinase 7/genetics , Protein Prenylation/drug effects , Protein Transport/drug effects , Quinolines/pharmacology , RNA Interference , Rosuvastatin Calcium/pharmacology , Signal Transduction/drug effects , Simvastatin/pharmacology , Tight Junctions/enzymology , Transfection , Zonula Occludens-1 Protein/metabolism
9.
J Biol Chem ; 292(31): 13034-13044, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28623232

ABSTRACT

Ion exchange in the renal tubules is fundamental to the maintenance of physiological ion levels. Claudin-16 (CLDN16) regulates the paracellular reabsorption of Mg2+ in the thick ascending limb of Henle's loop in the kidney, with dephosphorylation of CLDN16 increasing its intracellular distribution and decreasing paracellular Mg2+ permeability. CLDN16 is located in the tight junctions, but the mechanism regulating its localization is unclear. Using yeast two-hybrid systems, we found that CLDN16 binds to PDZRN3, a protein containing both RING-finger and PDZ domains. We also observed that the carboxyl terminus of the cytoplasmic CLDN16 region was required for PDZRN3 binding. PZDRN3 was mainly distributed in the cytosol of rat kidney cells and upon cell treatment with the protein kinase A inhibitor H-89, colocalized with CLDN16. H-89 also increased mono-ubiquitination and the association of CLDN16 with PDZRN3. Mono-ubiquitination levels of a K275A mutant were lower, and its association with PDZRN3 was reduced compared with wild-type (WT) CLDN16 and a K261A mutant, indicating that Lys-275 is the major ubiquitination site. An S217A mutant, a dephosphorylated form of CLDN16, localized to the cytosol along with PDZRN3 and the endosomal marker Rab7. PDZRN3 siRNA increased cell-surface localization of WT CLDN16 in H-89-treated cells or containing the S217A mutant and also suppressed CLDN16 endocytosis. Of note, H-89 decreased paracellular Mg2+ flux in WT CLDN16 cells, and PDZRN3 siRNA increased Mg2+ flux in the H-89-treated WT CLDN16 and S217A mutant cells. These results suggest that PDZRN3 mediates endocytosis of dephosphorylated CLDN16 and represents an important component of the CLDN16-trafficking machinery in the kidney.


Subject(s)
Claudins/metabolism , Endocytosis , Kidney Tubules/metabolism , Protein Processing, Post-Translational , Tight Junctions/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Substitution , Animals , Carrier Proteins/metabolism , Claudins/chemistry , Claudins/genetics , Dogs , Endocytosis/drug effects , Humans , Kidney Tubules/cytology , Kidney Tubules/drug effects , Lysine/metabolism , Madin Darby Canine Kidney Cells , Oligopeptides/genetics , Oligopeptides/metabolism , Phosphorylation/drug effects , Point Mutation , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , RNA Interference , Rats , Recombinant Fusion Proteins/metabolism , Tight Junctions/drug effects , Tight Junctions/enzymology , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitination/drug effects
10.
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
11.
Toxins (Basel) ; 8(9)2016 09 08.
Article in English | MEDLINE | ID: mdl-27618100

ABSTRACT

Deoxynivalenol (DON), produced by the plant pathogens Fusarium graminearum and Fusarium culmorum, is one of the most common mycotoxins, contaminating cereal and cereal-derived products. Although worldwide contamination of food and feed poses health threats to humans and animals, pigs are particularly susceptible to this mycotoxin. DON derivatives, such as deepoxy-deoxynivalenol (DOM-1), are produced by bacterial transformation of certain intestinal bacteria, which are naturally occurring or applied as feed additives. Intestinal epithelial cells are the initial barrier against these food- and feed-borne toxins. The present study confirms DON-induced activation of MAPK p44/42 and inhibition of p44/42 by MAPK-inhibitor U0126 monoethanolate. Influence of DON and DOM-1 on transepithelial electrical resistance (TEER), viability and expression of seven tight junction proteins (TJ), as well as the potential of U0126 to counteract DON-induced effects, was assessed. While DOM-1 showed no effect, DON significantly reduced TEER of differentiated IPEC-J2 and decreased expression of claudin-1 and -3, while leaving claudin-4; ZO-1, -2, and -3 and occludin unaffected. Inhibition of p44/42 counteracted DON-induced TEER decrease and restored claudin-3, but not claudin-1 expression. Therefore, effects of DON on TEER and claudin-3 are at least partially p44/42 mediated, while effects on viability and claudin-1 are likely mediated via alternative pathways.


Subject(s)
Epithelial Cells/drug effects , Intestinal Mucosa/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Tight Junctions/drug effects , Trichothecenes/toxicity , Animals , Calcium Signaling/drug effects , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Electric Impedance , Enzyme Activation , Epithelial Cells/enzymology , Epithelial Cells/pathology , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Permeability , Protein Kinase Inhibitors/pharmacology , Swine , Tight Junction Proteins/metabolism , Tight Junctions/enzymology , Tight Junctions/pathology , Time Factors
12.
Gene Ther ; 23(6): 489-99, 2016 06.
Article in English | MEDLINE | ID: mdl-26910760

ABSTRACT

Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are associated with diverse disorders and characterized by disruption of the alveolar-capillary barrier, leakage of edema fluid into the lung, and substantial inflammation leading to acute respiratory failure. Gene therapy is a potentially powerful approach to treat ALI/ARDS through repair of alveolar epithelial function. Herein, we show that delivery of a plasmid expressing ß1-subunit of the Na(+),K(+)-ATPase (ß1-Na(+),K(+)-ATPase) alone or in combination with epithelial sodium channel (ENaC) α1-subunit using electroporation not only protected from subsequent lipopolysaccharide (LPS)-mediated lung injury, but also treated injured lungs. However, transfer of α1-subunit of ENaC (α1-ENaC) alone only provided protection benefit rather than treatment benefit although alveolar fluid clearance had been remarkably enhanced. Gene transfer of ß1-Na(+),K(+)-ATPase, but not α1-ENaC, not only enhanced expression of tight junction protein zona occludins-1 (ZO-1) and occludin both in cultured cells and in mouse lungs, but also reduced pre-existing increase of lung permeability in vivo. These results demonstrate that gene transfer of ß1-Na(+),K(+)-ATPase upregulates tight junction formation and therefore treats lungs with existing injury, whereas delivery of α1-ENaC only maintains pre-existing tight junction but not for generation. This indicates that the restoration of epithelial/endothelial barrier function may provide better treatment of ALI/ARDS.


Subject(s)
Acute Lung Injury/genetics , Acute Lung Injury/therapy , Genetic Therapy/methods , Sodium-Potassium-Exchanging ATPase/genetics , Tight Junctions/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/enzymology , Animals , Disease Models, Animal , Electroporation/methods , Epithelial Sodium Channels/therapeutic use , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Plasmids/administration & dosage , Plasmids/genetics , Respiratory Distress Syndrome/therapy , Sodium-Potassium-Exchanging ATPase/administration & dosage , Tight Junctions/enzymology , Tight Junctions/genetics , Up-Regulation
13.
Am J Physiol Gastrointest Liver Physiol ; 309(12): G988-97, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26514773

ABSTRACT

Recent studies have implicated a pathogenic role for matrix metalloproteinases 9 (MMP-9) in inflammatory bowel disease. Although loss of epithelial barrier function has been shown to be a key pathogenic factor for the development of intestinal inflammation, the role of MMP-9 in intestinal barrier function remains unclear. The aim of this study was to investigate the role of MMP-9 in intestinal barrier function and intestinal inflammation. Wild-type (WT) and MMP-9(-/-) mice were subjected to experimental dextran sodium sulfate (DSS) colitis by administration of 3% DSS in drinking water for 7 days. The mouse colonic permeability was measured in vivo by recycling perfusion of the entire colon using fluorescently labeled dextran. The DSS-induced increase in the colonic permeability was accompanied by an increase in intestinal epithelial cell MMP-9 expression in WT mice. The DSS-induced increase in intestinal permeability and the severity of DSS colitis was found to be attenuated in MMP-9(-/-) mice. The colonic protein expression of myosin light chain kinase (MLCK) and phospho-MLC was found to be significantly increased after DSS administration in WT mice but not in MMP-9(-/-) mice. The DSS-induced increase in colonic permeability and colonic inflammation was attenuated in MLCK(-/-) mice and MLCK inhibitor ML-7-treated WT mice. The DSS-induced increase in colonic surface epithelial cell MLCK mRNA was abolished in MMP-9(-/-) mice. Lastly, increased MMP-9 protein expression was detected within the colonic surface epithelial cells in ulcerative colitis cases. These data suggest a role of MMP-9 in modulation of colonic epithelial permeability and inflammation via MLCK.


Subject(s)
Colitis/enzymology , Colon/enzymology , Dextran Sulfate , Intestinal Mucosa/enzymology , Matrix Metalloproteinase 9/metabolism , Tight Junctions/enzymology , Animals , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Colitis/prevention & control , Colon/drug effects , Colon/pathology , Disease Models, Animal , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Matrix Metalloproteinase 9/analysis , Matrix Metalloproteinase 9/deficiency , Matrix Metalloproteinase 9/genetics , Mice, Inbred C57BL , Mice, Knockout , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism , Permeability , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Severity of Illness Index , Signal Transduction , Tight Junction Proteins/metabolism , Tight Junctions/drug effects , Tight Junctions/pathology , Time Factors
14.
Int J Biochem Cell Biol ; 64: 120-5, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25757376

ABSTRACT

The establishment and maintenance of epithelial polarity must be correctly controlled for normal development and homeostasis. Tight junctions (TJ) in vertebrates define apical and basolateral membrane domains in polarized epithelia via bi-directional, complex signalling pathways between TJ themselves and the cytoskeleton they are associated with. RhoGTPases are central to these processes and evidence suggests that their regulation is coordinated by interactions between GEFs and GAPs with junctional, cytoplasmic adapter proteins. In this InFocus review we determine that the expression, localization or stability of a variety of these adaptor proteins is altered in various cancers, potentially representing an important mechanistic link between loss of polarity and cancer. We focus here, on two well characterized RhoGTPases Cdc42 and RhoA who's GEFs and GAPs are predominantly localized to TJ via cytoplasmic adaptor proteins.


Subject(s)
Neoplasms/enzymology , Tight Junctions/enzymology , rho GTP-Binding Proteins/physiology , Animals , Cell Adhesion Molecules/metabolism , Cell Polarity , Humans , Neoplasms/pathology , Signal Transduction
15.
J Lipid Res ; 56(3): 562-577, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25556764

ABSTRACT

To understand the mechanisms of 15(S)-HETE-induced endothelial cell (EC) barrier dysfunction, we examined the role of xanthine oxidase (XO). 15(S)-HETE induced junction adhesion molecule A (JamA) phosphorylation on Y164, Y218, and Y280 involving XO-mediated reactive oxygen species production and Src and Pyk2 activation, resulting in its dissociation from occludin, thereby causing tight junction (TJ) disruption, increased vascular permeability, and enhanced leukocyte and monocyte transmigration in vitro using EC monolayer and ex vivo using arteries as models. The phosphorylation of JamA on Y164, Y218, and Y280 appears to be critical for its role in 15(S)-HETE-induced EC barrier dysfunction, as mutation of any one of these amino acid residues prevented its dissociation from occludin and restored TJ integrity and barrier function. In response to high-fat diet (HFD) feeding, WT, but not 12/15-lipoxygenase (LO)(-/-), mice showed enhanced XO expression and its activity in the artery, which was correlated with increased aortic TJ disruption and barrier permeability with enhanced leukocyte adhesion and these responses were inhibited by allopurinol. These observations provide novel insights on the role of XO in 12/15-LO-induced JamA tyrosine phosphorylation and TJ disruption leading to increased vascular permeability in response to HFD.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Capillary Permeability/drug effects , Dietary Fats/adverse effects , Endothelium, Vascular/enzymology , Reactive Oxygen Species/metabolism , Tight Junctions/enzymology , Animals , Aorta/metabolism , Aorta/pathology , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Capillary Permeability/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Dietary Fats/pharmacology , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/genetics , Eicosapentaenoic Acid/metabolism , Endothelium, Vascular/pathology , Mice , Mice, Knockout , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Tight Junctions/genetics , Tight Junctions/pathology , Xanthine Oxidase/genetics , Xanthine Oxidase/metabolism
16.
Histochem Cell Biol ; 143(5): 471-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25511417

ABSTRACT

c-Jun N-terminal kinase (JNK), known as a stress-activated protein kinase, regulates normal epithelial biological processes, including assembly of adherens and tight junctions, and it is involved in the development of several cancers. The JNK inhibitor SP600125 enhances epithelial barrier function through modulation of tight junction molecules in normal human pancreatic epithelial cells. Furthermore, this JNK inhibitor suppresses the growth of human pancreatic cancer cells. However, the effects of SP600125 on the epithelial barrier in human pancreatic cancer cells remain unknown. In the present study, the JNK inhibitor SP600125 markedly enhanced the barrier function and cell elongation of well-differentiated human pancreatic cancer cell line HPAC in a Ca-switch model. The epithelial barrier function induced by SP600125 was regulated by phosphorylated ß-catenin without changes in the tight junction molecules. The cell elongation induced by SP600125 was closely related to the expression of the F-actin-binding protein DrebrinE. These findings suggest that JNK is involved in the regulation of the epithelial barrier function and cell shape during remodeling of pancreatic cancer cells. The JNK inhibitor SP600125 may have potential as a therapeutic drug for pancreatic cancer via induction of differentiation.


Subject(s)
Anthracenes/pharmacology , Calcium/metabolism , Cell Membrane Permeability/drug effects , Cell Shape/drug effects , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Tight Junctions/drug effects , Cell Line, Tumor , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphorylation , RNA Interference , Signal Transduction/drug effects , Tight Junctions/enzymology , Tight Junctions/pathology , Time Factors , Transfection , beta Catenin/metabolism
17.
Brain Struct Funct ; 220(1): 13-26, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24005261

ABSTRACT

Less disruption of the blood-brain barrier (BBB) after severe ischemic stroke is one of the beneficial outcomes of ischemic preconditioning (IP). However, the effect of IP on tight junctions (TJs), which regulate paracellular permeability of the BBB, is not well understood. In the present study, we examined IP-induced changes in TJs before and after middle cerebral artery occlusion (MCAO) in mice, and the association between changes in TJs and tolerance to a subsequent insult. After IP, we found decreased levels of transmembrane TJ proteins occludin and claudin-5, and widened gaps of TJs with perivascular swelling at the ultrastructural level in the brain. An inflammatory response was also observed. These changes were reversed by inhibition of extracellular signal-regulated kinase1/2 (ERK1/2) via the specific ERK1/2 inhibitor U0126. After MCAO, reduced brain edema and inflammatory responses were associated with altered levels of angiogenic factors and cytokines in preconditioned brains. Pretreatment with U0126 reversed the neuroprotective effects of IP against MCAO. These findings suggest that ERK1/2 activation has a pivotal role in IP-induced changes in TJs and inflammatory response, which serve to protect against BBB breakdown and inflammation after ischemic stroke.


Subject(s)
Infarction, Middle Cerebral Artery/pathology , Ischemic Preconditioning, Myocardial , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Tight Junctions/enzymology , Animals , Brain Edema/etiology , Brain Infarction/etiology , Butadienes/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Male , Mice , Mice, Inbred C57BL , Muscle Strength/drug effects , Neurologic Examination , Nitriles/pharmacology , Tight Junctions/drug effects , Tight Junctions/ultrastructure , Time Factors , Vascular Endothelial Growth Factor A/metabolism
18.
Int J Clin Exp Pathol ; 7(10): 7268-77, 2014.
Article in English | MEDLINE | ID: mdl-25400825

ABSTRACT

Diabetic retinopathy (DR) is a well-known serious complication of diabetes mellitus (DM), and can eventually advance to end-stage blindness. In the early stage of DR, endothelial cell barrier disorganized primarily and tight junction (TJ) protein composition transformed subsequently. The small GTPase RhoA and its downstream effector Rho-associated coiled-coil containing protein kinase 1 (ROCK1) regulate a mass of cellular processes, including cell adherence, proliferation, permeability and apoptosis. Although RhoA inhibitors have provided substantial clinical benefit as hypertonicity therapeutics, their use is limited by complex microenvironment as DR. While ample evidence indicates that TJ can be influenced by the RhoA/ROCK1 signaling, the underlying mechanisms remain incompletely understood. Here, we have uncovered a significant signaling network involved in diabetic retinal microvascular endothelial dysfunction (RMVED). Our results indicated that the activation of RhoA/ROCK1 pathway due to high glucose played a key role in microvascular endothelial cell dysfunction (MVED) by way of directly inducing TJ proteins over-expression during DR. We demonstrated that inhibition of RhoA/ROCK1 may attenuate the hypertonicity of endothelial cell caused by high glucose microenvironment meanwhile. Besides, chemical and pharmacological inhibitors of RhoA/ROCK1 pathway may partly block inflammation due to DR. Simultaneously, the apoptosis aroused by high glucose was also prevented considerably by fasudil, a kind of pharmacological inhibitor of RhoA/ROCK1 pathway. These findings indicate that RhoA/ROCK1 signaling directly modulates MVED, suggesting a novel therapeutic target for DR.


Subject(s)
Diabetic Retinopathy/etiology , Endothelial Cells/enzymology , Glucose/metabolism , Hyperglycemia/complications , Microvessels/enzymology , Retinal Vessels/enzymology , Signal Transduction , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Apoptosis , Capillary Permeability , Cell Line , Cellular Microenvironment , Diabetic Retinopathy/enzymology , Diabetic Retinopathy/pathology , Diabetic Retinopathy/physiopathology , Electric Impedance , Endothelial Cells/drug effects , Endothelial Cells/pathology , Hyperglycemia/enzymology , Macaca mulatta , Microvessels/drug effects , Microvessels/pathology , Microvessels/physiopathology , Protein Kinase Inhibitors/pharmacology , RNA Interference , Retinal Vessels/drug effects , Retinal Vessels/pathology , Retinal Vessels/physiopathology , Signal Transduction/drug effects , Tight Junctions/enzymology , Transfection , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics
19.
Thromb Haemost ; 112(2): 402-11, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24696092

ABSTRACT

Matriptase is a type-II transmembrane serine protease involved in epithelial homeostasis in both health and disease, and is implicated in the development and progression of a variety of cancers. Matriptase mediates its biological effects both via as yet undefined substrates and pathways, and also by proteolytic cleavage of a variety of well-defined protein substrates, several of which it shares with the closely-related protease hepsin. Development of targeted therapeutic strategies will require discrimination between these proteases. Here we have investigated cyclic microproteins of the squash Momordica cochinchinensis trypsin-inhibitor family (generated by total chemical synthesis) and found MCoTI-II to be a high-affinity (Ki 9 nM) and highly selective (> 1,000-fold) inhibitor of matriptase. MCoTI-II efficiently inhibited the proteolytic activation of pro-hepatocyte growth factor (HGF) by matriptase but not by hepsin, in both purified and cell-based systems, and inhibited HGF-dependent cell scattering. MCoTI-II also selectively inhibited the invasion of matriptase-expressing prostate cancer cells. Using a model of epithelial cell tight junction assembly, we also found that MCoTI-II could effectively inhibit the re-establishment of tight junctions and epithelial barrier function in MDCK-I cells after disruption, consistent with the role of matriptase in regulating epithelial integrity. Surprisingly, MCoTI-II was unable to inhibit matriptase-dependent proteolytic activation of prostasin, a GPI-anchored serine protease also implicated in epithelial homeostasis. These observations suggest that the unusually high selectivity afforded by MCoTI-II and its biological effectiveness might represent a useful starting point for the development of therapeutic inhibitors, and further highlight the role of matriptase in epithelial maintenance.


Subject(s)
Cyclotides/pharmacology , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Dogs , Electric Impedance , HEK293 Cells , Hepatocyte Growth Factor/metabolism , Humans , Madin Darby Canine Kidney Cells , Male , Molecular Targeted Therapy , Neoplasm Invasiveness , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Protein Precursors/metabolism , Serine Endopeptidases/genetics , Substrate Specificity , Tight Junctions/drug effects , Tight Junctions/enzymology , Time Factors , Transfection
20.
Exp Eye Res ; 122: 77-85, 2014 May.
Article in English | MEDLINE | ID: mdl-24667088

ABSTRACT

TNF-α induces loss of barrier integrity of the corneal endothelium through mechanisms involving the activation of p38 MAP kinase. This study has investigated the role of matrix metalloproteinase-9 (MMP-9), known to be activated by mechanisms downstream of p38 MAP kinase, on the breakdown of the barrier integrity. Experiments were performed with primary cultures of bovine corneal endothelium. Changes in the trans-endothelial electrical resistance (TER), a measure of barrier integrity, were measured by electric cell-substrate impedance sensing. The integrity of the apical junctional assembly was imaged by immunolocalization of ZO-1. MMP-9 activity in the conditioned medium of cells treated with TNF-α was visualized by gelatin zymography. Transcriptional activation of MMP-9 was assessed by real-time RT-PCR. Exposure to TNF-α led to significant disruption of ZO-1 and also caused a continuous decline in TER for more than 20 h. These effects were opposed by cycloheximide (protein synthesis inhibitor), GM-6001 (broad spectrum inhibitor of MMPs), minocycline (MMP-2 and MMP-9 inhibitor), and MMP-9 inhibitor I (selective MMP-9 inhibitor). Cycloheximide, GM-6001, and MMP-9 inhibitor I also attenuated the increase in permeability to FITC-dextran (10 kDa). In addition, TNF-α led to an increased MMP-9 activity in the conditioned medium as well as a nearly 20-fold increase in mRNA for MMP-9 but not for MMP-2. The functional activity and increase in mRNA levels of MMP-9 were blocked by SB-203580 (selective p38 MAP kinase inhibitor) and cycloheximide. In conclusion, transcriptional and translational activation of MMP-9, downstream of p38 MAP kinase signaling, is involved in the (TNF-α)-induced loss of corneal endothelial barrier integrity.


Subject(s)
Endothelium, Corneal/drug effects , Matrix Metalloproteinase 9/physiology , Tight Junctions/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cattle , Cells, Cultured , Cycloheximide/pharmacology , Dextrans/metabolism , Electric Impedance , Endothelium, Corneal/enzymology , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Gene Expression Regulation/physiology , Matrix Metalloproteinase Inhibitors/pharmacology , Protein Synthesis Inhibitors/pharmacology , Real-Time Polymerase Chain Reaction , Tight Junctions/enzymology , Zonula Occludens-1 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL