Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 756
Filter
1.
Am J Pathol ; 194(5): 673-683, 2024 05.
Article in English | MEDLINE | ID: mdl-38311119

ABSTRACT

Podocytes serve as part of the renal filtration unit with slit diaphragms. Although the structure of slit diaphragms between two cells is well characterized, how the tricellular contact of podocytes is organized and how it changes in injured podocytes remains unknown. This study focused on a tricellular junction protein, angulin-3, and its localization in healthy podocytes, in developmental stages, and in pathologic conditions, using a newly established monoclonal antibody. Angulin-3 was confined at tricellular junctions of primordial podocytes, then transiently localized at bicellular junctions as foot process interdigitation developed and the intercellular junctions rearranged into slit diaphragm, and eventually distributed in a sparse punctate pattern on the foot processes of adult podocytes. In the rodent podocyte injury models, angulin-3 showed bicellular localization between the foot processes, and the localization turned from punctate to dashed linear pattern along the effaced foot processes with the progression of podocyte injury. Angulin-3 also accumulated between foot processes in a linear pattern in kidney biopsy samples of human nephrotic syndrome. Additionally, the line length of angulin-3 staining signal correlated with risk of relapse under glucocorticoid therapy in patients with minimal change nephrotic syndrome. This study proposes an image program to score the linearity of the accumulation pattern of angulin-3 to evaluate the relapse risk of patients with minimal change nephrotic syndrome.


Subject(s)
Nephrosis, Lipoid , Podocytes , Adult , Humans , Podocytes/metabolism , Tight Junctions/pathology , Nephrosis, Lipoid/metabolism , Nephrosis, Lipoid/pathology , Intercellular Junctions/metabolism , Recurrence
2.
Am J Physiol Cell Physiol ; 327(2): C291-C309, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38826136

ABSTRACT

Per- and polyfluoroalkyl substances (PFASs) are a family of "forever chemicals" including perfluorooctane sulfonate (PFOS). These toxic chemicals do not break down in the environment or in our bodies. In the human body, PFOS and perfluoroctanoic acid (PFOA) have a half-life (T1/2) of about 4-5 yr so low daily consumption of these chemicals can accumulate in the human body to a harmful level over a long period. Although the use of PFOS in consumer products was banned in the United States in 2022/2023, this forever chemical remains detectable in our tap water and food products. Every American tested has a high level of PFAS in their blood (https://cleanwater.org/pfas-forever-chemicals). In this report, we used a Sertoli cell blood-testis barrier (BTB) model with primary Sertoli cells cultured in vitro with an established functional tight junction (TJ)-permeability barrier that mimicked the BTB in vivo. Treatment of Sertoli cells with PFOS was found to perturb the TJ-barrier, which was the result of cytoskeletal disruption across the cell cytoplasm, disrupting actin and microtubule polymerization. These changes thus affected the proper localization of BTB-associated proteins at the BTB. Using RNA-Seq transcriptome profiling, bioinformatics analysis, and pertinent biochemical and cell biology techniques, it was discovered that PFOS -induced Sertoli cell toxicity through the c-Jun N-terminal kinase (JNK; also known as stress-activated protein kinase, SAPK) and its phosphorylated/active form p-JNK signaling pathway. More importantly, KB-R7943 mesylate (KB), a JNK/p-JNK activator, was capable of blocking PFOS-induced Sertoli cell injury, supporting the notion that PFOS-induced cell injury can possibly be therapeutically managed.NEW & NOTEWORTHY PFOS induces Sertoli cell injury, including disruption of the 1) blood-testis barrier function and 2) cytoskeletal organization, which, in turn, impedes male reproductive function. These changes are mediated by JNK/p-JNK signaling pathway. However, the use of KB-R7943, a JNK/p-JNK activator was capable of blocking PFOS-induced Sertoli cell injury, supporting the possibility of therapeutically managing PFOS-induced reproductive dysfunction.


Subject(s)
Alkanesulfonic Acids , Fluorocarbons , JNK Mitogen-Activated Protein Kinases , Sertoli Cells , Fluorocarbons/toxicity , Alkanesulfonic Acids/toxicity , Male , Animals , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Sertoli Cells/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , RNA-Seq , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism , Tight Junctions/pathology , Cells, Cultured , Mice , Rats , Rats, Sprague-Dawley
3.
Am J Physiol Gastrointest Liver Physiol ; 327(2): G175-G187, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38860296

ABSTRACT

Accumulating literature suggests that the farnesoid-X receptor (FXR), a nuclear bile acid receptor best known for its role in bile acid homeostasis, is also a potent context-dependent regulator of inflammation. FXR may thus be relevant to several intestinal disease states including inflammatory bowel disease, necrotizing enterocolitis, and sepsis. In this study, we tested the effects of FXR deletion on acute murine intestinal inflammation. We found that FXR knockout (KO) mice were protected from intestinal injury and barrier dysfunction induced by lipopolysaccharide (LPS) injection, dithizone (DI)/Klebsiella, and cecal ligation/puncture models. In the LPS model, RNA sequencing and qPCR analysis showed that this protection correlated with substantial reduction in LPS-induced proinflammatory gene expression, including lower tissue levels of Il1a, Il1b, and Tnf. Examining functional effects on the epithelium, we found that LPS-induced tight junctional disruption as assessed by internalization of ZO-1 and occludin was ameliorated in FXR KO animals. Taken together, these data suggest a role for FXR in the intestinal barrier during inflammatory injury.NEW & NOTEWORTHY Intestinal barrier failure is a hallmark in gut-origin sepsis. We demonstrate that the intestinal barriers of farnesoid-X receptor (FXR) knockout (KO) animals are protected from inflammatory insult using multiple models of acute intestinal inflammation. This protection is due to decreased inflammatory cytokine production and maintenance of tight junctional architecture seen within the KO animals. This is the first report of FXR deletion being protective to the intestinal barrier.


Subject(s)
Intestinal Mucosa , Lipopolysaccharides , Mice, Knockout , Receptors, Cytoplasmic and Nuclear , Animals , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/deficiency , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Male , Inflammation/metabolism , Inflammation/genetics , Tight Junctions/metabolism , Tight Junctions/pathology , Disease Models, Animal
4.
Curr Opin Gastroenterol ; 40(2): 70-76, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38260939

ABSTRACT

PURPOSE OF REVIEW: In the pathophysiological context of cholangiopathies and more broadly of hepatopathies, while it is conceptually clear that the maintenance of inter-cholangiocyte and inter-hepatocyte tight junction integrity would be crucial for liver protection, only scarce studies have been devoted to this topic. Indeed, in the liver, alteration of tight junctions, the intercellular adhesion complexes that control paracellular permeability would result in leaky bile ducts and bile canaliculi, allowing bile reflux towards hepatic parenchyma, contributing to injury during the disease process. RECENT FINDINGS: Last decades have provided a great deal of information regarding both tight junction structural organization and signaling pathways related to tight junctions, providing clues about potential intervention to modulate paracellular permeability during cholangiopathies pathogenesis. Interestingly, several liver diseases have been reported to be associated with abnormal expression of one or several tight junction proteins. However, the question remains unanswered if these alterations would be primarily involved in the disease pathogenesis or if they would occur secondarily in the pathological course. SUMMARY: In this review, we provide an overview of tight junction disruptions described in various biliary diseases that should pave the way for defining new therapeutic targets in this field.


Subject(s)
Liver , Tight Junction Proteins , Humans , Tight Junction Proteins/metabolism , Liver/pathology , Bile Ducts , Tight Junctions/metabolism , Tight Junctions/pathology , Epithelial Cells
5.
Mol Biol Rep ; 51(1): 273, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38302794

ABSTRACT

Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by progressive neuronal damage and cognitive decline. Recent studies have shed light on the involvement of not only the blood-brain barrier (BBB) dysfunction but also significant alterations in cellular junctions in AD pathogenesis. In this review article, we explore the role of the BBB and cellular junctions in AD pathology, with a specific focus on the hippocampus. The BBB acts as a crucial protective barrier between the bloodstream and the brain, maintaining brain homeostasis and regulating molecular transport. Preservation of BBB integrity relies on various junctions, including gap junctions formed by connexins, tight junctions composed of proteins such as claudins, occludin, and ZO-1, as well as adherence junctions involving molecules like vascular endothelial (VE) cadherin, Nectins, and Nectin-like molecules (Necls). Abnormalities in these junctions and junctional components contribute to impaired neuronal signaling and increased cerebrovascular permeability, which are closely associated with AD advancement. By elucidating the underlying molecular mechanisms governing BBB and cellular junction dysfunctions within the context of AD, this review offers valuable insights into the pathogenesis of AD and identifies potential therapeutic targets for intervention.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Signal Transduction/physiology , Tight Junctions/metabolism , Tight Junctions/pathology
6.
Oral Dis ; 30(1): 3-22, 2024 Jan.
Article in English | MEDLINE | ID: mdl-36825434

ABSTRACT

Tight junctions (TJs) are cell-cell interactions that localize at the most apical portion of epithelial/endothelial cells. One of the predominant functions of TJs is to regulate material transport through paracellular pathway, which serves as a selective barrier. In recent years, the expression and function of TJs in salivary glands has attracted great interest. The characteristics of multiple salivary gland TJ proteins have been identified. During salivation, the activation of muscarinic acetylcholine receptor and transient receptor potential vanilloid subtype 1, as well as other stimuli, promote the opening of acinar TJs by inducing internalization of TJs, thereby contributing to increased paracellular permeability. Besides, endothelial TJs are also redistributed with leakage of blood vessels in cholinergic-stimulated submandibular glands. Furthermore, under pathological conditions, such as Sjögren's syndrome, diabetes mellitus, immunoglobulin G4-related sialadenitis, and autotransplantation, the integrity and barrier function of TJ complex are impaired and may contribute to hyposalivation. Moreover, in submandibular glands of Sjögren's syndrome mouse model and patients, the endothelial barrier is disrupted and involved in hyposecretion and lymphocytic infiltration. These findings enrich our understanding of the secretory mechanisms that link the importance of epithelial and endothelial TJ functions to salivation under both physiological and pathophysiological conditions.


Subject(s)
Sialorrhea , Sjogren's Syndrome , Mice , Animals , Humans , Tight Junctions/metabolism , Tight Junctions/pathology , Sjogren's Syndrome/pathology , Endothelial Cells , Salivary Glands/pathology , Saliva/metabolism , Submandibular Gland/metabolism
7.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article in English | MEDLINE | ID: mdl-34301883

ABSTRACT

Tuberous sclerosis complex 1 (Tsc1) is a tumor suppressor that functions together with Tsc2 to negatively regulate the mechanistic target of rapamycin complex 1 (mTORC1) activity. Here, we show that Tsc1 has a critical role in the tight junction (TJ) formation of epithelium, independent of its role in Tsc2 and mTORC1 regulation. When an epithelial cell establishes contact with neighboring cells, Tsc1, but not Tsc2, migrates from the cytoplasm to junctional membranes, in which it binds myosin 6 to anchor the perijunctional actin cytoskeleton to ß-catenin and ZO-1. In its absence, perijunctional actin cytoskeleton fails to form. In mice, intestine-specific or inducible, whole-body Tsc1 ablation disrupts adherens junction/TJ structures in intestine or skin epithelia, respectively, causing Crohn's disease-like symptoms in the intestine or psoriasis-like phenotypes on the skin. In patients with Crohn's disease or psoriasis, junctional Tsc1 levels in epithelial tissues are markedly reduced, concomitant with the TJ structure impairment, suggesting that Tsc1 deficiency may underlie TJ-related diseases. These findings establish an essential role of Tsc1 in the formation of cell junctions and underpin its association with TJ-related human diseases.


Subject(s)
Actin Cytoskeleton/metabolism , Crohn Disease/pathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Psoriasis/pathology , Tight Junctions/pathology , Tuberous Sclerosis Complex 1 Protein/metabolism , Tuberous Sclerosis Complex 1 Protein/physiology , Actin Cytoskeleton/genetics , Animals , Case-Control Studies , Crohn Disease/genetics , Crohn Disease/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mice, Knockout , Psoriasis/genetics , Psoriasis/metabolism , Signal Transduction , Tight Junctions/genetics , Tight Junctions/metabolism , Tuberous Sclerosis Complex 1 Protein/genetics
8.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33443222

ABSTRACT

Effective therapies for alcohol-associated liver disease (ALD) are limited; therefore, the discovery of new therapeutic agents is greatly warranted. Toll-like receptor 7 (TLR7) is a pattern recognition receptor for single-stranded RNA, and its activation prevents liver fibrosis. We examined liver and intestinal damage in Tlr7-/- mice to determine the role of TLR7 in ALD pathogenesis. In an alcoholic hepatitis (AH) mouse model, hepatic steatosis, injury, and inflammation were induced by chronic binge ethanol feeding in mice, and Tlr7 deficiency exacerbated these effects. Because these results demonstrated that endogenous TLR7 signaling activation is protective in the AH mouse model, we hypothesized that TLR7 activation may be an effective therapeutic strategy for ALD. Therefore, we investigated the therapeutic effect of TLR7 agonistic agent, 1Z1, in the AH mouse model. Oral administration of 1Z1 was well tolerated and prevented intestinal barrier disruption and bacterial translocation, which thus suppressed ethanol-induced hepatic injury, steatosis, and inflammation. Furthermore, 1Z1 treatment up-regulated the expression of antimicrobial peptides, Reg3b and Reg3g, in the intestinal epithelium, which modulated the microbiome by decreasing and increasing the amount of Bacteroides and Lactobacillus, respectively. Additionally, 1Z1 up-regulated intestinal interleukin (IL)-22 expression. IL-22 deficiency abolished the protective effects of 1Z1 in ethanol-induced liver and intestinal damage, suggesting intestinal IL-22 as a crucial mediator for 1Z1-mediated protection in the AH mouse model. Collectively, our results indicate that TLR7 signaling exerts protective effects in the AH mouse model and that a TLR7 ligand, 1Z1, holds therapeutic potential for the treatment of AH.


Subject(s)
Ethanol/toxicity , Interleukins/metabolism , Intestinal Mucosa/metabolism , Liver Diseases, Alcoholic/drug therapy , Membrane Glycoproteins/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 7/metabolism , Administration, Oral , Animals , Bacteroides/drug effects , Disease Models, Animal , Fatty Liver/complications , Fatty Liver/genetics , Fatty Liver/metabolism , Female , Gastrointestinal Microbiome/drug effects , Inflammation/complications , Inflammation/genetics , Inflammation/metabolism , Intestinal Mucosa/drug effects , Lactobacillus/drug effects , Ligands , Liver Diseases, Alcoholic/genetics , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/physiopathology , Membrane Glycoproteins/agonists , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs , Pancreatitis-Associated Proteins/genetics , Pancreatitis-Associated Proteins/metabolism , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Signal Transduction/genetics , Tight Junctions/drug effects , Tight Junctions/pathology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/genetics , Interleukin-22
9.
Eur Heart J ; 44(14): 1248-1261, 2023 04 07.
Article in English | MEDLINE | ID: mdl-36638776

ABSTRACT

AIMS: Whether changes in endothelial tight junctions (TJs) lead to the formation of thoracic aortic aneurysm and dissection (TAAD) and serve as an early indicator and therapeutic target remains elusive. METHODS AND RESULTS: Single-cell RNA sequencing analysis showed aberrant endothelial TJ expressions in the thoracic aortas of patients with TAAD. In a ß-aminopropionitrile (BAPN)-induced TAAD mouse model, endothelial TJ function was disrupted in the thoracic aortas at an early stage (5 and 10 days) as observed by a vascular permeability assay, while the intercellular distribution of crucial TJ components was significantly decreased by en face staining. For the non-invasive detection of endothelial TJ function, two dextrans of molecular weights 4 and 70 kDa were conjugated with the magnetic resonance imaging (MRI) contrast agent Gd-DOTA to synthesize FITC-dextran-DOTA-Gd and rhodamine B-dextran-DOTA-Gd. MRI images showed that both probes accumulated in the thoracic aortas of the BAPN-fed mice. Particularly, the mice with increased accumulated signals from 5 to 10 days developed TAAD at 14 days, whereas the mice with similar signals between the two time points did not. Furthermore, the protease-activated receptor 2 inhibitor AT-1001, which seals TJs, alleviated the BAPN-induced impairment of endothelial TJ function and expression and subsequently reduced TAAD incidence. Notably, endothelial-targeted ZO-1 conditional knockout increased TAAD incidence. Mechanistically, vascular inflammation and edema were observed in the thoracic aortas of the BAPN-fed mice, whereas these phenomena were attenuated by AT-1001. CONCLUSION: The disruption of endothelial TJ function is an early event prior to TAAD formation, herein serving as a potential indicator and a promising target for TAAD.


Subject(s)
Aortic Aneurysm, Thoracic , Aortic Dissection , Mice , Animals , Aminopropionitrile/adverse effects , Tight Junctions/metabolism , Tight Junctions/pathology , Signal Transduction , Aortic Aneurysm, Thoracic/prevention & control
10.
Gastroenterology ; 162(3): 844-858, 2022 03.
Article in English | MEDLINE | ID: mdl-34822802

ABSTRACT

BACKGROUND & AIMS: The protozoa Giardia duodenalis is a major cause of gastrointestinal illness worldwide, but underlying pathophysiological mechanisms remain obscure, partly due to the absence of adequate cellular models. We aimed at overcoming these limitations and recapitulating the authentic series of pathogenic events in the primary human duodenal tissue by using the human organoid system. METHODS: We established a compartmentalized cellular transwell system with electrophysiological and barrier properties akin to duodenal mucosa and dissected the events leading to G. duodenalis-induced barrier breakdown by functional analysis of transcriptional, electrophysiological, and tight junction components. RESULTS: Organoid-derived cell layers of different donors showed a time- and parasite load-dependent leak flux indicated by collapse of the epithelial barrier upon G. duodenalis infection. Gene set enrichment analysis suggested major expression changes, including gene sets contributing to ion transport and tight junction structure. Solute carrier family 12 member 2 and cystic fibrosis transmembrane conductance regulator-dependent chloride secretion was reduced early after infection, while changes in the tight junction composition, localization, and structural organization occurred later as revealed by immunofluorescence analysis and freeze fracture electron microscopy. Functionally, barrier loss was linked to the adenosine 3',5'-cyclic monophosphate (cAMP)/protein kinase A-cAMP response element-binding protein signaling pathway. CONCLUSIONS: Data suggest a previously unknown sequence of events culminating in intestinal barrier dysfunction upon G. duodenalis infection during which alterations of cellular ion transport were followed by breakdown of the tight junctional complex and loss of epithelial integrity, events involving a cAMP/protein kinase A-cAMP response element-binding protein mechanism. These findings and the newly established organoid-derived model to study G. duodenalis infection may help to explore new options for intervening with disease and infection, in particular relevant for chronic cases of giardiasis.


Subject(s)
Giardiasis/physiopathology , Intestinal Mucosa/physiopathology , Ion Transport , Signal Transduction , Tight Junctions/physiology , Apoptosis , Caco-2 Cells , Chlorides/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Duodenum , Electric Impedance , Giardia lamblia , Giardiasis/genetics , Giardiasis/immunology , Humans , Interleukin-1/genetics , Ion Transport/genetics , NF-kappa B/genetics , Organoids , Parasite Load , Solute Carrier Family 12, Member 2/genetics , Tight Junctions/genetics , Tight Junctions/pathology , Tight Junctions/ultrastructure , Transcriptome , Tumor Necrosis Factor-alpha/genetics
11.
Hepatology ; 75(1): 89-103, 2022 01.
Article in English | MEDLINE | ID: mdl-34392560

ABSTRACT

BACKGROUND AND AIMS: Biliary atresia is a severe inflammatory and fibrosing cholangiopathy of neonates of unknown etiology. The onset of cholestasis at birth implies a prenatal onset of liver dysfunction. Our aim was to investigate the mechanisms linked to abnormal cholangiocyte development. APPROACH AND RESULTS: We generated biliary organoids from liver biopsies of infants with biliary atresia and normal and diseased controls. Organoids emerged from biliary atresia livers and controls and grew as lumen-containing spheres with an epithelial lining of cytokeratin-19pos albuminneg SOX17neg cholangiocyte-like cells. Spheres had similar gross morphology in all three groups and expressed cholangiocyte-enriched genes. In biliary atresia, cholangiocyte-like cells lacked a basal positioning of the nucleus, expressed fewer developmental and functional markers, and displayed misorientation of cilia. They aberrantly expressed F-actin, ß-catenin, and Ezrin, had low signals for the tight junction protein zonula occludens-1 (ZO-1), and displayed increased permeability as evidenced by a higher Rhodamine-123 (R123) signal inside organoids after verapamil treatment. Biliary atresia organoids had decreased expression of genes related to EGF signaling and FGF2 signaling. When treated with EGF+FGF2, biliary atresia organoids expressed differentiation (cytokeratin 7 and hepatocyte nuclear factor 1 homeobox B) and functional (somatostatin receptor 2, cystic fibrosis transmembrane conductance regulator [CFTR], aquaporin 1) markers, restored polarity with improved localization of F-actin, ß-catenin and ZO-1, increased CFTR function, and decreased uptake of R123. CONCLUSIONS: Organoids from biliary atresia are viable and have evidence of halted epithelial development. The induction of developmental markers, improved cell-cell junction, and decreased epithelial permeability by EGF and FGF2 identifies potential strategies to promote epithelial maturation and function.


Subject(s)
Bile Ducts/pathology , Biliary Atresia/pathology , Cholestasis/pathology , Epithelial Cells/pathology , Organoids/pathology , Adolescent , Bile Ducts/cytology , Bile Ducts/growth & development , Biliary Atresia/complications , Biopsy , Case-Control Studies , Cells, Cultured , Child , Child, Preschool , Cholestasis/etiology , Epithelial Cells/cytology , Healthy Volunteers , Humans , Infant , Infant, Newborn , Primary Cell Culture , Tight Junctions/pathology
12.
PLoS Biol ; 18(11): e3000946, 2020 11.
Article in English | MEDLINE | ID: mdl-33253145

ABSTRACT

Inflammation of the central nervous system (CNS) induces endothelial blood-brain barrier (BBB) opening as well as the formation of a tight junction barrier between reactive astrocytes at the Glia Limitans. We hypothesized that the CNS parenchyma may acquire protection from the reactive astrocytic Glia Limitans not only during neuroinflammation but also when BBB integrity is compromised in the resting state. Previous studies found that astrocyte-derived Sonic hedgehog (SHH) stabilizes the BBB during CNS inflammatory disease, while endothelial-derived desert hedgehog (DHH) is expressed at the BBB under resting conditions. Here, we investigated the effects of endothelial Dhh on the integrity of the BBB and Glia Limitans. We first characterized DHH expression within endothelial cells at the BBB, then demonstrated that DHH is down-regulated during experimental autoimmune encephalomyelitis (EAE). Using a mouse model in which endothelial Dhh is inducibly deleted, we found that endothelial Dhh both opens the BBB via the modulation of forkhead box O1 (FoxO1) transcriptional activity and induces a tight junctional barrier at the Glia Limitans. We confirmed the relevance of this glial barrier system in human multiple sclerosis active lesions. These results provide evidence for the novel concept of "chronic neuroinflammatory tolerance" in which BBB opening in the resting state is sufficient to stimulate a protective barrier at the Glia Limitans that limits the severity of subsequent neuroinflammatory disease. In summary, genetic disruption of the BBB generates endothelial signals that drive the formation under resting conditions of a secondary barrier at the Glia Limitans with protective effects against subsequent CNS inflammation. The concept of a reciprocally regulated CNS double barrier system has implications for treatment strategies in both the acute and chronic phases of multiple sclerosis pathophysiology.


Subject(s)
Blood-Brain Barrier/physiology , Blood-Brain Barrier/physiopathology , Adherens Junctions/pathology , Adherens Junctions/physiology , Animals , Antigens, CD/genetics , Antigens, CD/physiology , Astrocytes/pathology , Astrocytes/physiology , Cadherins/genetics , Cadherins/physiology , Capillary Permeability/genetics , Capillary Permeability/physiology , Claudin-5/genetics , Claudin-5/physiology , Down-Regulation , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Endothelial Cells/pathology , Endothelial Cells/physiology , Female , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Hedgehog Proteins/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Neuroglia/pathology , Neuroglia/physiology , Tight Junctions/pathology , Tight Junctions/physiology
13.
Mol Biol Rep ; 50(3): 2007-2014, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36536183

ABSTRACT

BACKGROUND: Celiac disease (CD) is a hereditary immune-mediated disorder, which is along with the enormous production of pro-inflammatory cytokines and the reduced level of tight junction proteins. The aim of this study was to determine the expression of TNF-α, IFN-γ, IL-18, Occludin, miR-122-5p and miR-197-3p genes in duodenal biopsies of treated CD patients in comparison to the controls. METHODS AND RESULTS: Biopsy specimens were taken from the duodenum of 50 treated CD patients (36 (72%) females and 14 (28%) males with mean age of 37.06 ± 7.02 years) and 50 healthy controls (17 (34%) females and 33 (66%) males with mean age of 34.12 ± 4.9). Total RNA was isolated, cDNA was synthesized and mRNA expression of TNF-α, IFN-γ, IL-18, Occludin, miR-122-5p and miR-197-3p were quantified by relative qPCR using B2M and U6 as internal control genes. All data were evaluated using SPSS (V.21) and GraphPad Prism (V.5). Our results showed that there was no significant difference between patients and controls for intestinal mRNA expression of TNF-α, IFN-γ, IL-18, Occludin, and miR-122-5p (p > 0.05) and the expression of miR-197-3p was significantly increased in CD patients relative to control subjects (p = 0.049). CONCLUSION: This study suggests that adherence to GFD may have a positive effect on the tight junction (TJ) permeability and in this process, miR-197-3p plays an important role. Increased expression of miR-197-3p with a final protective effect on Occludin expression can be further studied as a complement therapeutic target for Celiac disease.


Subject(s)
Celiac Disease , MicroRNAs , Adult , Female , Humans , Male , Celiac Disease/genetics , Celiac Disease/pathology , Diet, Gluten-Free , Interleukin-18/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Occludin/genetics , Permeability , RNA, Messenger/metabolism , Tight Junctions/genetics , Tight Junctions/metabolism , Tight Junctions/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
14.
Dig Dis Sci ; 68(12): 4306-4320, 2023 12.
Article in English | MEDLINE | ID: mdl-37773554

ABSTRACT

The intestinal barrier is composed of several essential elements including luminal enzymes, bile acids, water layer, epithelial layer, and enterocyte layer. It acts as a dynamic interface between the luminal contents of food, commensal and pathogenic bacteria, and the gastrointestinal tract. The role of barrier dysfunction is of significant research interest in the development and targeted treatment of chronic inflammatory gastrointestinal conditions, such as inflammatory bowel disease. This review aims to examine the role of intestinal barrier dysfunction in the development of inflammatory bowel disease, the pathophysiology of increased barrier permeability in inflammatory bowel disease, and to explore potential treatment targets and clinical applications.


Subject(s)
Gastrointestinal Diseases , Inflammatory Bowel Diseases , Humans , Intestinal Mucosa/pathology , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/pathology , Intestines/pathology , Permeability , Tight Junctions/pathology
15.
Alzheimers Dement ; 19(11): 5185-5197, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37166032

ABSTRACT

INTRODUCTION: Vascular amyloid beta (Aß) protein deposits were detected in retinas of mild cognitively impaired (MCI) and Alzheimer's disease (AD) patients. We tested the hypothesis that the retinal vascular tight junctions (TJs) were compromised and linked to disease status. METHODS: TJ components and Aß expression in capillaries and larger blood vessels were determined in post mortem retinas from 34 MCI or AD patients and 27 cognitively normal controls and correlated with neuropathology. RESULTS: Severe decreases in retinal vascular zonula occludens-1 (ZO-1) and claudin-5 correlating with abundant arteriolar Aß40 deposition were identified in MCI and AD patients. Retinal claudin-5 deficiency was closely associated with cerebral amyloid angiopathy, whereas ZO-1 defects correlated with cerebral pathology and cognitive deficits. DISCUSSION: We uncovered deficiencies in blood-retinal barrier markers for potential retinal imaging targets of AD screening and monitoring. Intense retinal arteriolar Aß40 deposition suggests a common pathogenic mechanism of failed Aß clearance via intramural periarterial drainage.


Subject(s)
Alzheimer Disease , Cerebral Amyloid Angiopathy , Retina , Humans , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Cerebral Amyloid Angiopathy/pathology , Claudin-5/metabolism , Tight Junctions/metabolism , Tight Junctions/pathology , Retina/pathology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology
16.
Clin Gastroenterol Hepatol ; 20(5): 1019-1028.e3, 2022 05.
Article in English | MEDLINE | ID: mdl-34607017

ABSTRACT

BACKGROUND & AIMS: In addition to gastric sensorimotor dysfunctions, functional dyspepsia (FD) is also variably associated with duodenal micro-inflammation and epithelial barrier dysfunction, the pathogenesis and clinical significance of which are unknown. Our hypothesis was that miRNAs and/or inflammation degrade epithelial barrier proteins, resulting in increased duodenal mucosal permeability in FD. METHODS: We compared the duodenal mucosal gene expression and miRNAs, in vivo permeability (lactulose-mannitol excretion between 0 and 60 and 60 and 120 minutes after saccharide ingestion), ex vivo assessments (transmucosal resistance, fluorescein isothiocyanate [FITC]-dextran flux, and basal ion transport), and duodenal histology (light and electron microscopy) in 40 patients with FD and 24 controls. RESULTS: Compared with controls, the mRNA expression of several barrier proteins (zonula occludens-1, occludin, claudin-12, and E-cadherin) was modestly reduced (ie, a fold change of 0.8-0.85) in FD with increased expression of several miRNAs (eg, miR-142-3p and miR-144-3-p), which suppress these genes. The urinary lactulose excretion and the lactulose:mannitol ratio between 60 and 120 minutes were greater in FD than in controls (P < .05). The FITC-dextran flux, which reflects paracellular permeability, was inversely correlated (r = -0.32, P = .03) with transmucosal resistance and directly correlated (r = 0.4, P = .02) with lactulose:mannitol ratio. Other parameters (mucosal eosinophils, intraepithelial lymphocytes, and mast cells, transmucosal resistance, FITC-dextran flux, average intercellular distance, and proportion of dilated junctions) were not significantly different between groups. CONCLUSIONS: In FD, there is a modest reduction in the expression of several duodenal epithelial barrier proteins, which may be secondary to up-regulation of regulatory miRNAs, and increased small intestinal permeability measured in vivo.


Subject(s)
Dyspepsia , MicroRNAs , Dyspepsia/pathology , Humans , Inflammation/pathology , Intestinal Mucosa/pathology , Lactulose , Mannitol/metabolism , MicroRNAs/genetics , Permeability , Tight Junctions/metabolism , Tight Junctions/pathology
17.
Gastroenterology ; 160(4): 1240-1255.e3, 2021 03.
Article in English | MEDLINE | ID: mdl-33189700

ABSTRACT

BACKGROUND & AIMS: The down-regulated in adenoma (DRA) protein, encoded by SLC26A3, a key intestinal chloride anion exchanger, has recently been identified as a novel susceptibility gene for inflammatory bowel disease (IBD). However, the mechanisms underlying the increased susceptibility to inflammation induced by the loss of DRA remain elusive. Compromised barrier is a key event in IBD pathogenesis. The current studies were undertaken to elucidate the impact of DRA deficiency on epithelial barrier integrity and to define underlying mechanisms. METHODS: Wild-type and DRA-knockout (KO) mice and crypt-derived colonoids were used as models for intestinal epithelial response. Paracellular permeability was measured by using fluorescein isothiocyanate-dextran flux. Immunoblotting, immunofluorescence, immunohistochemistry, and ribonucleoprotein immunoprecipitation assays were performed. Gut microbiome analysis was conducted to investigate the impact of DRA deficiency on gut microbial communities. RESULTS: DRA-KO mice exhibited an increased colonic paracellular permeability with significantly decreased levels of tight junction/adherens junction proteins, including ZO-1, occludin, and E-cadherin. A similar expression pattern of occludin and E-cadherin was observed in colonoids derived from DRA-KO mice and short hairpin RNA-mediated DRA knockdown in Caco-2 cells. Microbial analysis showed gut dysbiosis in DRA-KO mice. However, cohousing studies showed that dysbiosis played only a partial role in maintaining tight junction protein expression. Furthermore, our results showed increased binding of RNA-binding protein CUGBP1 with occludin and E-cadherin genes in DRA-KO mouse colon, suggesting that posttranscriptional mechanisms play a key role in gut barrier dysfunction. CONCLUSIONS: To our knowledge, our studies demonstrate a novel role of DRA in maintaining the intestinal epithelial barrier function and potential implications of its dysregulation in IBD pathogenesis.


Subject(s)
Antiporters/deficiency , Chloride-Bicarbonate Antiporters/deficiency , Dysbiosis/immunology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Sulfate Transporters/deficiency , Animals , Antiporters/genetics , CELF1 Protein/metabolism , Caco-2 Cells , Cadherins/metabolism , Chloride-Bicarbonate Antiporters/genetics , Disease Models, Animal , Dysbiosis/microbiology , Dysbiosis/pathology , Gene Knockdown Techniques , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Male , Mice , Mice, Knockout , Occludin/metabolism , Permeability , Sulfate Transporters/genetics , Tight Junctions/pathology
18.
Am J Pathol ; 191(5): 872-884, 2021 05.
Article in English | MEDLINE | ID: mdl-33607043

ABSTRACT

Defective intestinal tight junction (TJ) barrier is an important pathogenic factor of inflammatory bowel disease. To date, no effective therapies that specifically target the intestinal TJ barrier are available. The purpose of this study was to identify probiotic bacterial species or strains that induce a rapid and sustained enhancement of intestinal TJ barrier and protect against the development of intestinal inflammation by targeting the TJ barrier. After high-throughput screening of >20 Lactobacillus and other probiotic bacterial species or strains, a specific strain of Lactobacillus acidophilus, referred to as LA1, uniquely produced a marked enhancement of the intestinal TJ barrier. LA1 attached to the apical membrane surface of intestinal epithelial cells in a Toll-like receptor (TLR)-2-dependent manner and caused a rapid increase in enterocyte TLR-2 membrane expression and TLR-2/TLR-1 and TLR-2/TLR-6 hetero-complex-dependent enhancement in intestinal TJ barrier function. Oral administration of LA1 caused a rapid enhancement in mouse intestinal TJ barrier, protected against a dextran sodium sulfate (DSS) increase in intestinal permeability, and prevented the DSS-induced colitis in a TLR-2- and intestinal TJ barrier-dependent manner. In conclusion, we report for the first time that a specific strain of LA causes a strain-specific enhancement of intestinal TJ barrier through a novel mechanism that involves the TLR-2 receptor complex and protects against the DSS-induced colitis by targeting the intestinal TJ barrier.


Subject(s)
Colitis/prevention & control , Inflammation/prevention & control , Lactobacillus acidophilus/physiology , Probiotics , Toll-Like Receptor 2/metabolism , Animals , Colitis/chemically induced , Colitis/microbiology , Colitis/pathology , Dextran Sulfate/adverse effects , Epithelial Cells/pathology , Intestines/drug effects , Intestines/pathology , Mice , Mice, Inbred C57BL , Permeability/drug effects , Tight Junctions/drug effects , Tight Junctions/pathology , Toll-Like Receptor 2/genetics
19.
Exp Eye Res ; 220: 109094, 2022 07.
Article in English | MEDLINE | ID: mdl-35490836

ABSTRACT

Diabetic retinopathy is a major cause of blindness in developed countries, and is characterized by deterioration of barrier function causing vascular hyperpermeability and retinal edema. Vascular endothelial growth factor (VEGF) is a major mediator of diabetic macular edema. Although anti-VEGF drugs are the first-line treatment for diabetic macular edema, some cases are refractory to anti-VEGF therapy. Osteopontin (OPN) is a phosphoglycoprotein with diverse functions and expressed in various cells and tissues. Elevated OPN level has been implicated in diabetic retinopathy, but whether OPN is involved in hyperpermeability remains unclear. Using streptozotocin-induced diabetic mice (STZ mice) and human retinal endothelial cells (HRECs), we tested the hypothesis that up-regulated OPN causes tight junction disruption, leading to vascular hyperpermeability. The serum and retinal OPN concentrations were elevated in STZ mice compared to controls. Intravitreal injection of anti-OPN neutralizing antibody (anti-OPN Ab) suppressed vascular hyperpermeability and prevented decreases in claudin-5 and ZO-1 gene expression levels in the retina of STZ mice. Immunohistochemical staining of retinal vessels in STZ mice revealed claudin-5 immunoreactivity with punctate distribution and attenuated ZO-1 immunoreactivity, and these changes were prevented by anti-OPN Ab. Intravitreal injection of anti-OPN Ab did not change VEGF gene expression or protein concentration in retina of STZ mice. In an in vitro study, HRECs were exposed to normal glucose or high glucose with or without OPN for 48 h, and barrier function was evaluated by transendothelial electrical resistance and Evans blue permeation. Barrier function deteriorated under high glucose condition, and was further exacerbated by the addition of OPN. Immunofluorescence localization of claudin-5 and ZO-1 demonstrated punctate appearance with discontinuous junction in HRECs exposed to high glucose and OPN. There were no changes in VEGF and VEGF receptor-2 expression levels in HRECs by exposure to OPN. Our results suggest that OPN induces tight junction disruption and vascular hyperpermeability under diabetic conditions. Targeting OPN may be an effective approach to manage diabetic retinopathy.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Retinopathy , Macular Edema , Osteopontin , Tight Junctions , Animals , Blood-Retinal Barrier , Claudin-5/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/metabolism , Endothelial Cells/metabolism , Glucose/pharmacology , Macular Edema/metabolism , Mice , Osteopontin/genetics , Osteopontin/metabolism , Retina/metabolism , Retinal Vessels/metabolism , Streptozocin , Tight Junctions/metabolism , Tight Junctions/pathology , Vascular Endothelial Growth Factor A/metabolism
20.
FASEB J ; 35(2): e21189, 2021 02.
Article in English | MEDLINE | ID: mdl-33200449

ABSTRACT

Disassembly of tight junctions is a major cause of intestinal barrier dysfunction under total parenteral nutrition (TPN), but the precise mechanisms have not been fully understood. Normally, RNA binding protein Lin 28A is highly restricted to embryonic stem cells and dramatically decreases as differentiation progresses; however, in our preliminary study it was found aberrantly increased in the intestinal epithelial cells of TPN rats, and thus its mechanism of action needs to be addressed. Herein, we report a pivotal role of Lin 28A in the regulation of tight junctions, which induces a sustained translational repression of Occludin, leading to disruption of intestinal barrier function under TPN. Using a rat model of TPN, we found time-dependent upregulation of Lin 28A, negatively correlated with Occludin. Using mouse intestinal organoids and human gut-derived Caco-2 cells as in vitro models, we found that expression of Occludin could be significantly suppressed by ectopic overexpression of Lin 28A. The underlying mechanisms may be partially attributed to translational repression, as the abundance of Occludin transcripts in polysomes was dramatically reduced by Lin 28A (polysomal profiling). Furthermore, Lin 28A was found to directly bind to Occludin mRNA 3' untranslated coding region (UTR), thereby repressing the translation of Occludin transcripts through decapping enzyme 1A (DCP1a). Taken together, our findings revealed that Lin 28A/Occludin axis may be a novel mechanism accounting for the development of barrier dysfunction under TPN.


Subject(s)
Enterocytes/metabolism , Occludin/metabolism , Parenteral Nutrition/adverse effects , RNA-Binding Proteins/metabolism , Tight Junctions/metabolism , Animals , Caco-2 Cells , Cells, Cultured , Enterocytes/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Signal Transduction , Tight Junctions/pathology
SELECTION OF CITATIONS
SEARCH DETAIL