Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.813
Filter
1.
J Tradit Chin Med ; 44(5): 885-895, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39380219

ABSTRACT

OBJECTIVE: To investigate the mechanism of the protective effect of modified Pulsatilla decoction (, MPD) on the mechanical barrier of the ulcerative colitis (UC) intestinal epithelium in vitro and in vivo. METHODS: We established an intestinal epithelial crypt cell line-6 cell barrier injury model by using lipopolysaccharide (LPS). The model was then treated with p38 mitogen-activated protein kinase-myosin light chain kinase (p38MAPK-MLCK) pathway inhibitors, p38MAPK-MLCK pathway silencing genes (si-p38MAPK, si-NF-κB, and si-MLCK), and MPD respectively. Transepithelial electronic resistance (TEER) measurements and permeability assays were performed to assess barrier function. Immunofluorescence staining of tight junctions (TJ) was performed. In in vivo experiment, dextran sodium sulfate-induced colitis rat model was conducted to evaluate the effect of MPD and mesalazine on UC. The rats were scored using the disease activity index based on their clinical symptoms. Transmission electron microscopy and hematoxylin-eosin staining were used to examine morphological changes in UC rats. Western blotting and real-time quantitative polymerase chain reaction were performed to examine the gene and protein expression of significant differential molecules. RESULTS: In in vitro study, LPS-induced intestinal barrier dysfunction was inhibited by p38MAPK-MLCK pathway inhibitors and p38MAPK-MLCK pathway gene silencing. Silencing of p38MAPK-MLCK pathway genes decreased TJ expression. MPD treatment partly restored the LPS-induced decreased in TEER and increase in permeability. MPD increased the gene and protein expression of TJ, while down-regulated the LPS-induced high expression of p-p38MAPK and p-MLC. In UC model rats, MPD could ameliorate body weight loss and disease activity index, relieve colonic pathology, up-regulate TJ expression as well as decrease the expression of p-p38MAPK and p-MLC in UC rat colonic mucosal tissue. CONCLUSIONS: The p38MAPK-MLCK signaling pathway can affect mechanical barrier function and TJ expression in the intestinal epithelium. MPD restores TJ expression and attenuates intestinal epithelial barrier damage by suppressing the p38MAPK-MLCK pathway.


Subject(s)
Colitis, Ulcerative , Drugs, Chinese Herbal , Intestinal Mucosa , Myosin-Light-Chain Kinase , p38 Mitogen-Activated Protein Kinases , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/genetics , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/chemically induced , Rats , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/administration & dosage , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Humans , Rats, Sprague-Dawley , Signal Transduction/drug effects , Cell Line , Tight Junctions/drug effects , Tight Junctions/metabolism
2.
J Cell Sci ; 137(19)2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39350674

ABSTRACT

SGEF (also known as ARHGEF26), a RhoG specific GEF, can form a ternary complex with the Scribble polarity complex proteins Scribble and Dlg1, which regulates the formation and maintenance of adherens junctions and barrier function of epithelial cells. Notably, silencing SGEF results in a dramatic downregulation of both E-cadherin and ZO-1 (also known as TJP1) protein levels. However, the molecular mechanisms involved in the regulation of this pathway are not known. Here, we describe a novel signaling pathway governed by the Scribble-SGEF-Dlg1 complex. Our results show that the three members of the ternary complex are required to maintain the stability of the apical junctions, ZO-1 protein levels and tight junction (TJ) permeability. In contrast, only SGEF is necessary to regulate E-cadherin levels. The absence of SGEF destabilizes the E-cadherin-catenin complex at the membrane, triggering a positive feedback loop that exacerbates the phenotype through the repression of E-cadherin transcription in a process that involves the internalization of E-cadherin by endocytosis, ß-catenin signaling and the transcriptional repressor Slug (also known as SNAI2).


Subject(s)
Cadherins , Epithelial Cells , Membrane Proteins , Rho Guanine Nucleotide Exchange Factors , Snail Family Transcription Factors , Zonula Occludens-1 Protein , Cadherins/metabolism , Cadherins/genetics , Humans , Epithelial Cells/metabolism , Zonula Occludens-1 Protein/metabolism , Zonula Occludens-1 Protein/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Rho Guanine Nucleotide Exchange Factors/genetics , Animals , Discs Large Homolog 1 Protein/metabolism , Discs Large Homolog 1 Protein/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Transcription, Genetic , Transcription Factors/metabolism , Transcription Factors/genetics , Madin Darby Canine Kidney Cells , Tight Junctions/metabolism , Dogs , Signal Transduction , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Protein Stability , beta Catenin/metabolism , beta Catenin/genetics
3.
FASEB J ; 38(18): e70049, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39275889

ABSTRACT

In this study, we have explored the role of the KATNB1 gene, a microtubule-severing protein, in the seminiferous epithelium of the rat testis. Our data have shown that KATNB1 expressed in rat brain, testes, and Sertoli cells. KATNB1 was found to co-localize with α-tubulin showing a unique stage-specific distribution across the seminiferous epithelium. Knockdown of KATNB1 by RNAi led to significant disruption of the tight junction (TJ) permeability barrier function in primary Sertoli cells cultured in vitro with an established functional TJ-barrier, as well as perturbations in the microtubule and actin cytoskeleton organization. The disruption in these cytoskeletal structures, in turn, led to improper distribution of TJ and basal ES proteins essential for maintaining the Sertoli TJ function. More importantly, overexpression of KATNB1 in the testis in vivo was found to block cadmium-induced blood-testis barrier (BTB) disruption and testis injury. KATNB1 exerted its promoting effects on BTB and spermatogenesis through corrective spatiotemporal expression of actin- and microtubule-based regulatory proteins by maintaining the proper organization of cytoskeletons in the testis, illustrating its plausible therapeutic implication. In summary, Katanin regulatory subunit B1 (KATNB1) plays a crucial role in BTB and spermatogenesis through its effects on the actin- and microtubule-based cytoskeletons in Sertoli cells and testis, providing important insights into male reproductive biology.


Subject(s)
Blood-Testis Barrier , Katanin , Sertoli Cells , Animals , Male , Sertoli Cells/metabolism , Rats , Katanin/metabolism , Katanin/genetics , Blood-Testis Barrier/metabolism , Cytoskeleton/metabolism , Rats, Sprague-Dawley , Tight Junctions/metabolism , Spermatogenesis/physiology , Cells, Cultured , Seminiferous Epithelium/metabolism , Testis/metabolism , Microtubules/metabolism , Tubulin/metabolism
4.
Biosens Bioelectron ; 266: 116721, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39226753

ABSTRACT

Live-cell label-free imaging of a microscopic biological barrier, generally referred to as 'tight junction', was realized by a recently developed electric-double-layer modulation imaging (EDLMI). The method allowed quantitative imaging of barrier integrity in real time, thus being an upper compatible of transepithelial electrical resistance (TEER) which is a conventional standard technique to evaluate spatially averaged barrier integrity. We demonstrate that the quantitative and real-time imaging capability of EDLMI unveils fundamental dynamics of biological barrier, some of which are totally different from conventional understandings.


Subject(s)
Biosensing Techniques , Humans , Biosensing Techniques/methods , Tight Junctions/metabolism , Electric Impedance
5.
J Cell Sci ; 137(18)2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39319625

ABSTRACT

Cingulin (CGN) tethers nonmuscle myosin 2B (NM2B; heavy chain encoded by MYH10) to tight junctions (TJs) to modulate junctional and apical cortex mechanics. Here, we studied the role of the CGN-nonmuscle myosin 2 (NM2) interaction in epithelial morphogenesis and nanoscale organization of CGN by expressing wild-type and mutant CGN constructs in CGN-knockout Madin-Darby canine kidney (MDCK) epithelial cells. We show that the NM2-binding region of CGN is required to promote normal cyst morphogenesis of MDCK cells grown in three dimensions and to maintain the C-terminus of CGN in a distal position with respect to the ZO-2 (or TJP2)-containing TJ submembrane region, whereas the N-terminus of CGN is localized more proximal to the TJ membrane. We also show that the CGN mutant protein that causes deafness in human and mouse models is localized at TJs but does not bind to NM2B, resulting in decreased TJ membrane tortuosity. These results indicate that the interaction between CGN and NM2B regulates epithelial tissue morphogenesis and nanoscale organization of CGN and suggest that CGN regulates the auditory function of hair cells by organizing the actomyosin cytoskeleton to modulate the mechanics of the apical and junctional cortex.


Subject(s)
Morphogenesis , Nonmuscle Myosin Type IIB , Dogs , Animals , Madin Darby Canine Kidney Cells , Nonmuscle Myosin Type IIB/metabolism , Nonmuscle Myosin Type IIB/genetics , Tight Junctions/metabolism , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Humans , Epithelial Cells/metabolism , Protein Binding , Epithelium/metabolism , Epithelium/growth & development , Mice
6.
Clin Sci (Lond) ; 138(19): 1209-1226, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39222031

ABSTRACT

Allergens and Th2 cytokines affect the homeostatic environment in the airways, leading to increased mucus production by goblet cells associated with altered adherens junctional complex (AJC) and tight junction (TJ) proteins responsible for maintaining epithelial barrier function. Circadian clock-dependent regulatory mechanisms such as inflammation and epithelial barrier function are gaining more attention due to their therapeutic potential against allergic inflammatory lung diseases. Currently, there are no studies to support whether REV-ERBα activation can attenuate Th2 cytokine-induced epithelial barrier dysfunction in human bronchial epithelial cells. We hypothesized that Th2 cytokine-induced epithelial barrier dysfunction may be protected by activating REV-ERBα. Treatment with Th2 cytokines or HDM significantly reduced the cell impedance, as confirmed by transepithelial electrical resistance (TEER). However, pre-treatment with SR10067 attenuated Th2 cytokine-induced barrier dysfunction, such as decreased permeability, improved TEER, localization of AJC and TJ proteins, and mRNA and protein levels of selected epithelial barrier and circadian clock targets. Overall, we showed for the first time that REV-ERBα activation regulates altered epithelial barrier function that may have direct implications for the treatment of asthma and other allergic diseases.


Subject(s)
Bronchi , Cytokines , Epithelial Cells , Nuclear Receptor Subfamily 1, Group D, Member 1 , Th2 Cells , Humans , Nuclear Receptor Subfamily 1, Group D, Member 1/agonists , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Cytokines/metabolism , Bronchi/drug effects , Bronchi/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism , Tight Junctions/metabolism , Tight Junctions/drug effects , Electric Impedance , Thiophenes/pharmacology , Adherens Junctions/drug effects , Adherens Junctions/metabolism
7.
Genes (Basel) ; 15(9)2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39336792

ABSTRACT

Blood-testis barrier (BTB) genes are crucial for the cellular mechanisms of spermatogenesis as they protect against detrimental cytotoxic agents, chemicals, and pathogens, thereby maintaining a sterile environment necessary for sperm development. BTB proteins predominantly consist of extensive tight and gap junctions formed between Sertoli cells. These junctions form a crucial immunological barrier restricting the intercellular movement of substances and molecules within the adluminal compartment. Epithelial tight junctions are complex membrane structures composed of various integral membrane proteins, including claudins, zonula occludens-1, and occludin. Inter-testicular cell junction proteins undergo a constant process of degradation and renewal. In addition, the downregulation of genes crucial to the development and preservation of cell junctions could disrupt the functionality of the BTB, potentially leading to male infertility. Oxidative stress and inflammation may contribute to disrupted spermatogenesis, resulting in male infertility. L-cysteine is a precursor to glutathione, a crucial antioxidant that helps mitigate damage and inflammation resulting from oxidative stress. Preclinical research indicates that L-cysteine may offer protective benefits against testicular injury and promote the expression of BTB genes. This review emphasizes various BTB genes essential for preserving its structural integrity and facilitating spermatogenesis and male fertility. Furthermore, it consolidates various research findings suggesting that L-cysteine may promote the expression of BTB-associated genes, thereby aiding in the maintenance of testicular functions.


Subject(s)
Blood-Testis Barrier , Cysteine , Spermatogenesis , Male , Blood-Testis Barrier/metabolism , Humans , Animals , Cysteine/metabolism , Tight Junctions/metabolism , Oxidative Stress , Infertility, Male/genetics , Infertility, Male/metabolism , Sertoli Cells/metabolism , Sertoli Cells/drug effects , Testis/metabolism
8.
Gut Microbes ; 16(1): 2409247, 2024.
Article in English | MEDLINE | ID: mdl-39349383

ABSTRACT

The anaerobic spirochete Brachyspira causes intestinal spirochetosis, characterized by the intimate attachment of bacterial cells to the colonic mucosa, potentially leading to symptoms such as diarrhea, abdominal pain, and weight loss. Despite the clinical significance of Brachyspira infections, the mechanism of the interaction between Brachyspira and the colon epithelium is not known. We characterized the molecular mechanism of the B. pilosicoli-epithelium interaction and its impact on the epithelial barrier during infection. Through a proteomics approach, we identified BPP43_05035 as a candidate B. pilosicoli surface protein that mediates bacterial attachment to cultured human colonic epithelial cells. The crystal structure of BPP43_05035 revealed a globular lipoprotein with a six-bladed beta-propeller domain. Blocking the native BPP43_05035 on B. pilosicoli, either with a specific antibody or via competitive inhibition, abrogated its binding to epithelial cells, which required cell surface-exposed N-glycans. Proximity labeling and interaction assays revealed that BPP43_05035 bound to tight junctions, thereby increasing the permeability of the epithelial monolayer. Extending our investigation to humans, we discovered a downregulation of tight junction and brush border genes in B. pilosicoli-infected patients carrying detectable levels of epithelium-bound BPP43_05035. Collectively, our findings identify BPP43_05035 as a B. pilosicoli adhesin that weakens the colonic epithelial barrier during infection.


Subject(s)
Adhesins, Bacterial , Bacterial Adhesion , Brachyspira , Epithelial Cells , Intestinal Mucosa , Humans , Adhesins, Bacterial/metabolism , Adhesins, Bacterial/genetics , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Brachyspira/metabolism , Brachyspira/genetics , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Colon/microbiology , Colon/metabolism , Gram-Negative Bacterial Infections/microbiology , Tight Junctions/metabolism , Tight Junctions/microbiology
9.
Sci Total Environ ; 952: 175886, 2024 Nov 20.
Article in English | MEDLINE | ID: mdl-39218115

ABSTRACT

The study evaluates the impact of environmental toxicants, such as polycyclic aromatic hydrocarbons (PAHs), on circadian regulations and functions of brain endothelial cells, which form the main structural element of the blood-brain barrier (BBB). PAH are lipophilic and highly toxic environmental pollutants that accumulate in human and animal tissues. Environmental factors related to climate change, such as an increase in frequency and intensity of wildfires or enhanced strength of hurricanes or tropical cyclones, may lead to redistribution of these toxicants and enhanced human exposure. These natural disasters are also associated with disruption of circadian rhythms in affected populations, linking increased exposure to environmental toxicants to alterations of circadian rhythm pathways. Several vital physiological processes are coordinated by circadian rhythms, and disruption of the circadian clock can contribute to the development of several diseases. The blood-brain barrier (BBB) is crucial for protecting the brain from blood-borne harmful substances, and its integrity is influenced by circadian rhythms. Exposure of brain endothelial cells to a human and environmentally-relevant PAH mixture resulted in dose-dependent alterations of expression of critical circadian modulators, such as Clock, Bmal1, Cry1/2, and Per1/2. Moreover, silencing of the circadian Clock gene potentiated the impact of PAHs on the expression of the main tight junction genes and proteins (namely, claudin-5, occludin, JAM-2, and ZO-2), as well as mitochondrial bioenergetics. Findings from this study contribute to a better understanding of pathological influence of PAH-induced health effects, especially those related to circadian rhythm disruption.


Subject(s)
Blood-Brain Barrier , Circadian Rhythm , Polycyclic Aromatic Hydrocarbons , Tight Junctions , Polycyclic Aromatic Hydrocarbons/toxicity , Humans , Blood-Brain Barrier/drug effects , Tight Junctions/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Energy Metabolism/drug effects , Endothelial Cells/drug effects , Endothelial Cells/physiology , Environmental Pollutants/toxicity
10.
Article in English | MEDLINE | ID: mdl-39276851

ABSTRACT

Regulation of salt and water balance occupies a dominant role in the physiology of many animals and often relies on the function of the renal system. In the mammalian kidney, epithelial ion and water transport requires high degree of coordination between the transcellular and paracellular pathways, the latter being defined by the intercellular tight junctions (TJs). TJs seal the paracellular pathway in a highly specialized manner, either by forming a barrier against the passage of solutes and/or water or by allowing the passage of ions and/or water through them. This functional TJ plasticity is now known to be provided by the members of the claudin family of tetraspan proteins. Unlike mammalian nephron, the renal structures of insects, the Malpighian tubules, lack TJs and instead have smooth septate junctions (sSJs) as paracellular barrier forming junctions. Many questions regarding the molecular and functional properties of sSJs remain open but research on model species have begun to inform our understanding. The goal of this commentary is to highlight key concepts and most recent findings that have emerged from the molecular and functional dissection of paracellular barriers in the mammalian and insect renal epithelia.


Subject(s)
Kidney , Tight Junctions , Animals , Tight Junctions/metabolism , Tight Junctions/physiology , Kidney/physiology , Kidney/metabolism , Humans , Epithelial Cells/metabolism , Epithelial Cells/physiology , Claudins/metabolism , Malpighian Tubules/metabolism , Malpighian Tubules/physiology , Epithelium/physiology , Epithelium/metabolism
11.
Redox Biol ; 76: 103359, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39298837

ABSTRACT

Intestinal permeabilization is central to the pathophysiology of chronic gut inflammation. This study investigated the efficacy of glucoraphanin (GR), prevalent in cruciferous vegetables, particularly broccoli, and its derivative sulforaphane (SF), in inhibiting tumor necrosis factor alpha (TNFα)-induced Caco-2 cell monolayers inflammation and permeabilization through the regulation of redox-sensitive events. TNFα binding to its receptor led to a rapid increase in oxidant production and subsequent elevation in the mRNA levels of NOX1, NOX4, and Duox2. GR and SF dose-dependently mitigated both these short- and long-term alterations in redox homeostasis. Downstream, GR and SF inhibited the activation of the redox-sensitive signaling cascades NF-κB (p65 and IKK) and MAPK ERK1/2, which contribute to inflammation and barrier permeabilization. GR (1 µM) and SF (0.5-1 µM) prevented TNFα-induced monolayer permeabilization and the associated reduction in the levels of the tight junction (TJ) proteins occludin and ZO-1. Both GR and SF also mitigated TNFα-induced increased mRNA levels of the myosin light chain kinase, which promotes TJ opening. Molecular docking suggests that although GR is mostly not absorbed, it could interact with extracellular and membrane sites in NOX1. Inhibition of NOX1 activity by GR would mitigate TNFα receptor downstream signaling and associated events. These findings support the concept that not only SF, but also GR, could exert systemic health benefits by protecting the intestinal barrier against inflammation-induced permeabilization, in part by regulating redox-sensitive pathways. GR has heretofore not been viewed as a biologically active molecule, but rather, the benign precursor of highly active SF. The consumption of GR and/or SF-rich vegetables or supplements in the diet may offer a means to mitigate the detrimental consequences of intestinal permeabilization, not only in disease states but also in conditions characterized by chronic inflammation of dietary and lifestyle origin.


Subject(s)
Glucosinolates , Imidoesters , Inflammation , Isothiocyanates , Oximes , Sulfoxides , Tumor Necrosis Factor-alpha , Humans , Sulfoxides/pharmacology , Isothiocyanates/pharmacology , Caco-2 Cells , Tumor Necrosis Factor-alpha/metabolism , Oximes/pharmacology , Imidoesters/pharmacology , Imidoesters/metabolism , Glucosinolates/pharmacology , Inflammation/metabolism , Inflammation/drug therapy , Signal Transduction/drug effects , Tight Junctions/metabolism , Tight Junctions/drug effects , Permeability/drug effects , Cell Membrane Permeability/drug effects , Oxidation-Reduction/drug effects , NADPH Oxidases/metabolism , NADPH Oxidases/genetics , NF-kappa B/metabolism
12.
Mediators Inflamm ; 2024: 1484806, 2024.
Article in English | MEDLINE | ID: mdl-39262415

ABSTRACT

Background: Colitis is a refractory intestinal inflammatory disease significantly affecting the quality of a patient's life and increasing the risk of exacerbation. The primary factors leading to colitis encompass infections, insufficient blood flow, and the buildup of collagen as well as white blood cells. Among various available therapeutics, 5-methoxytryptophan (5-MTP) has emerged as one of the protectants by inhibiting inflammatory damage. Nonetheless, there is no report on the role of 5-MTP in the treatment of colitis. Materials and Methods: To verify the anti-inflammatory effect of 5-MTP in vivo, we first constructed mouse model with dextran sulfate sodium-induced colitis. Furthermore, the macrophage infiltration and release of inflammatory factors through western blot (WB) and hematoxylin-eosin staining analyses were examined. Intestinal epithelial cell tight junction damage and apoptosis were investigated by WB analysis, immunofluorescence, and terminal deoxynucleotidyl transferase dUTP nick end labeling staining. Finally, we examined the generation of cellular inflammation and analyzed the influence of 5-MTP on M1 polarization at the cellular level. Results: This study initially confirmed that 5-MTP possessed an excellent therapeutic effect on colitis. 5-MTP inhibits macrophage infiltration and the generation of inflammatory factors. In addition to its effects on immune cells, 5-MTP significantly inhibits intestinal epithelial cell tight junction damage and apoptosis in vivo. Moreover, it inhibits inflammation and M1 polarization response in vitro. Conclusion: 5-MTP counteracts excessive inflammation, thereby preventing intestinal epithelial tight junction damage. In addition, inhibition of apoptosis suggests that 5-MTP may be a potential therapeutic agent for colitis.


Subject(s)
Colitis , Dextran Sulfate , Intestinal Mucosa , Mice, Inbred C57BL , Tryptophan , Animals , Dextran Sulfate/toxicity , Colitis/chemically induced , Colitis/drug therapy , Mice , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Tryptophan/analogs & derivatives , Tryptophan/pharmacology , Inflammation/drug therapy , Male , Apoptosis/drug effects , Macrophages/drug effects , Macrophages/metabolism , Humans , Disease Models, Animal , Tight Junctions/drug effects , Tight Junctions/metabolism
13.
Nat Cancer ; 5(9): 1371-1389, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39289595

ABSTRACT

Carcinomas are associated with metastasis to specific organs while sparing others. Breast cancer presents with lung metastasis but rarely kidney metastasis. Using this difference as an example, we queried the mechanism(s) behind the proclivity for organ-specific metastasis. We used spontaneous and implant models of metastatic mammary carcinoma coupled with inflammatory tissue fibrosis, single-cell sequencing analyses and functional studies to unravel the causal determinants of organ-specific metastasis. Here we show that lung metastasis is facilitated by angiopoietin 2 (Ang2)-mediated suppression of lung-specific endothelial tight junction protein Claudin 5, which is augmented by the inflammatory fibrotic microenvironment and prevented by anti-Ang2 blocking antibodies, while kidney metastasis is prevented by non-Ang2-responsive Claudins 2 and 10. Suppression of Claudins 2 and 10 was sufficient to induce the emergence of kidney metastasis. This study illustrates the influence of organ-specific vascular heterogeneity in determining organotropic metastasis, independent of cancer cell-intrinsic mechanisms.


Subject(s)
Claudins , Kidney Neoplasms , Lung Neoplasms , Tight Junctions , Animals , Female , Mice , Claudins/metabolism , Claudins/genetics , Tight Junctions/metabolism , Kidney Neoplasms/pathology , Kidney Neoplasms/metabolism , Humans , Lung Neoplasms/secondary , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Tumor Microenvironment , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Cell Line, Tumor , Neoplasm Metastasis
14.
Am J Physiol Gastrointest Liver Physiol ; 327(4): G545-G557, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39104325

ABSTRACT

Increased intestinal permeability is a manifestation of cystic fibrosis (CF) in people with CF (pwCF) and in CF mouse models. CF transmembrane conductance regulator knockout (Cftr KO) mouse intestine exhibits increased proliferation and Wnt/ß-catenin signaling relative to wild-type mice (WT). Since the Rho GTPase Cdc42 plays a central role in intestinal epithelial proliferation and tight junction remodeling, we hypothesized that Cdc42 may be altered in the Cftr KO crypts. Immunofluorescence showed distinct tight junction localization of Cdc42 in Cftr KO fresh crypts and enteroids, the latter indicating an epithelial-autonomous feature. Quantitative PCR and immunoblots revealed similar expression of Cdc42 in the Cftr KO crypts/enteroids relative to WT, whereas pulldown assays showed increased GTP-bound (active) Cdc42 in proportion to total Cdc42 in Cftr KO enteroids. Cdc42 activity in the Cftr KO and WT enteroids could be reduced by inhibition of the Wnt transducer Disheveled. With the use of a dye permeability assay, Cftr KO enteroids exhibited increased paracellular permeability to 3 kDa dextran relative to WT. Leak permeability and Cdc42 tight junction localization were reduced to a greater extent by inhibition of Wnt/ß-catenin signaling with endo-IWR1 in Cftr KO relative to WT enteroids. Increased proliferation or inhibition of Cdc42 activity with ML141 in WT enteroids had no effect on permeability. In contrast, inhibition of Cdc42 with ML141 increased permeability to both 3 kDa dextran and tight junction impermeant 500 kDa dextran in Cftr KO enteroids. These data suggest that increased constitutive Cdc42 activity may alter the stability of paracellular permeability in Cftr KO crypt epithelium.NEW & NOTEWORTHY Increased tight junction localization and GTP-bound activity of the Rho GTPase Cdc42 was identified in small intestinal crypts and enteroids of cystic fibrosis (CF) transmembrane conductance regulator knockout (Cftr KO) mice. The increase in epithelial Cdc42 activity was associated with increased Wnt signaling. Paracellular flux of an uncharged solute (3 kDa dextran) in Cftr KO enteroids indicated a moderate leak permeability under basal conditions that was strongly exacerbated by Cdc42 inhibition. These findings suggest increased activity of Cdc42 in the Cftr KO intestine underlies alterations in intestinal permeability.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator , Intestinal Mucosa , Tight Junctions , cdc42 GTP-Binding Protein , Animals , Mice , cdc42 GTP-Binding Protein/metabolism , cdc42 GTP-Binding Protein/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Mice, Knockout , Permeability , Tight Junctions/metabolism , Wnt Signaling Pathway
15.
Exp Eye Res ; 247: 110062, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39187056

ABSTRACT

Exposure to VEGF-A165a over several days leads to a persistent dysfunction of the very tight barrier formed by immortalized endothelial cells of the bovine retina (iBREC). Elevated permeability of the barrier is indicated by low cell index values determined by electric cell-substrate impedance measurements, by lower amounts of claudin-1, and by disruption of the homogenous and continuous staining of vascular endothelial cadherin at the plasma membrane. Because of findings that suggest modulation of VEGF-A's detrimental effects on the inner blood-retina barrier by the angiogenic growth factor angiopoietin-2, we investigated in more detail in vitro whether this growth factor indeed changes the stability of the barrier formed by retinal endothelial cells or modulates effects of VEGF-A. In view of the clinical relevance of anti-VEGF therapy, we also studied whether blocking VEGF-A-driven signaling is sufficient to prevent barrier dysfunction induced by a combination of both growth factors. Although angiopoietin-2 stimulated proliferation of iBREC, the formed barrier was not weakened at a concentration of 3 nM: Cell index values remained high and expression or subcellular localization of claudin-1 and vascular endothelial cadherin, respectively, were not affected. Angiopoietin-2 enhanced the changes induced by VEGF-A165a and this was more pronounced at lower concentrations of VEGF-A165a. Specific inhibition of the VEGF receptors with tivozanib as well as interfering with binding of VEGF-A to its receptors with bevacizumab prevented the detrimental effects of the growth factors; dual binding of angiopoietin-2 and VEGF-A by faricimab was marginally more efficient. Uptake of extracellular angiopoietin-2 by iBREC can be efficiently prevented by addition of faricimab which is also internalized by the cells. Exposure of the cells to faricimab over several days stabilized their barrier, confirming that inhibition of VEGF-A signaling is not harmful to this cell type. Taken together, our results confirm the dominant role of VEGF-A165a in processes resulting in increased permeability of retinal endothelial cells in which angiopoietin-2 might play a minor modulating role.


Subject(s)
Angiopoietin-2 , Blood-Retinal Barrier , Cadherins , Cell Proliferation , Vascular Endothelial Growth Factor A , Animals , Cattle , Angiogenesis Inhibitors/pharmacology , Angiopoietin-2/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cell Proliferation/drug effects , Cells, Cultured , Claudin-1/metabolism , Electric Impedance , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/cytology , Peptide Fragments , Retinal Vessels/cytology , Retinal Vessels/metabolism , Tight Junctions/metabolism , Vascular Endothelial Growth Factor A/metabolism
16.
Food Chem Toxicol ; 192: 114966, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39197527

ABSTRACT

Advanced glycation end products (AGEs) are a spectrum of complex compounds widely found in processed foods and frequently consumed by humans. AGEs are implicated in impairing the intestinal barrier, but the underlying mechanisms remain unclear. This study investigated the effects of three types of AGEs on gene expression of tight junctions (TJs) in colorectal epithelial HT-29 cells, and observed minimal alterations in TJs expression. Given the important role of subepithelial macrophages in regulating the intestinal barrier, we explored whether AGEs affect the intestinal barrier via the involvement of macrophages. Notably, a significant downregulation of TJs expression was observed when supernatants from AGEs-treated RAW264.7 macrophage cells were transferred to HT-29 cells. Further investigations indicated that AGEs increased IL-6 levels in RAW264.7 cells, subsequently triggering STAT3 activation and suppressing TJs expression in HT-29 cells. The role of STAT3 activation was confirmed by observing enhanced TJs expression in HT-29 cells following pretreatment with an inhibitor of STAT3 activation prior to the transfer of the conditioned medium. These findings demonstrated that AGEs impaired the intestinal barrier via macrophage-mediated STAT3 activation, shedding light on the mechanisms underlying AGEs-induced intestinal barrier injury and related food safety risks.


Subject(s)
Glycation End Products, Advanced , Intestinal Mucosa , Macrophages , STAT3 Transcription Factor , STAT3 Transcription Factor/metabolism , Glycation End Products, Advanced/metabolism , Humans , Animals , Macrophages/drug effects , Macrophages/metabolism , Mice , HT29 Cells , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , RAW 264.7 Cells , Tight Junctions/drug effects , Tight Junctions/metabolism , Interleukin-6/metabolism , Interleukin-6/genetics
17.
Life Sci ; 355: 122990, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39154812

ABSTRACT

Cytotoxic metabolites originating from the peripheral circulation can induce central nervous system complications associated with diabetes. Since a large proportion of these metabolites bind to plasma albumin, mechanisms for transporting albumin-metabolite complexes into the brain exist under diabetic conditions. Secreted protein acidic and rich in cysteine (SPARC) is one of the vesicular transport receptors responsible for albumin transport. This study aimed to investigate the changes in SPARC expression and cellular albumin transfer under high-glucose conditions and evaluate the permeability of molecules with high protein-bound properties to the brain tissue. Glucose (30 mM) increased SPARC expression, and intracellular albumin accumulation in NIH3T3 cells. In addition, these changes were observed in the brain of ob/ob mice. Brain microvessels function as a physiological barrier to limit the penetration of molecules from the peripheral blood circulation into the brain by forming tight junctions. Although protein expression of molecules involved in tight junction formation and cell adhesion was increased in the brain microvessels of ob/ob mice, molecular transfer into the brain through cellular junctions was not enhanced. However, Evans blue dye injected into the peripheral vein and endogenous advanced glycation end-products, exerted a high protein-binding property and accumulated in their brains. These observations indicate that peripheral molecules with high protein-binding properties invade the brain tissue and bind to albumin through transcytosis mediated by SPARC.


Subject(s)
Brain , Microvessels , Osteonectin , Animals , Osteonectin/metabolism , Mice , Brain/metabolism , Brain/blood supply , Microvessels/metabolism , Male , NIH 3T3 Cells , Albumins/metabolism , Glucose/metabolism , Biological Transport , Blood-Brain Barrier/metabolism , Glycation End Products, Advanced/metabolism , Mice, Obese , Transcytosis , Mice, Inbred C57BL , Tight Junctions/metabolism
19.
Phytomedicine ; 133: 155953, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39154527

ABSTRACT

BACKGROUND: Mycoplasma gallisepticum (MG) has long been a pathogenic microorganism threatening the global poultry industry. Previous studies have demonstrated that the mechanism by which quercetin (QUE) inhibits the colonization of MG in chicks differs from that of antibiotics. However, the molecular mechanism by which QUE facilitates the clearance of MG remains unclear. PURPOSE: The aim of this study was to investigate the molecular mechanism of MG clearance by QUE, with the expectation of providing new options for the treatment of MG. METHODS: A model of MG infection in chicks and MG-induced M1 polarization in HD-11 cells were established. The mechanism of QUE clearance of MG was investigated by evaluating the relationship between tracheal mucosal barrier integrity, antibody levels, Th1/Th2 immune balance and macrophage metabolism and M1/M2 polarization balance. Furthermore, network pharmacology and molecular docking techniques were employed to explore the potential molecular pathways connecting QUE, M2 polarization, and fatty acid oxidation (FAO). RESULTS: The findings indicate that QUE remodels tracheal mucosal barrier function by regulating tight junctions and secretory immunoglobulin A (sIgA) expression levels. This process entails the regulatory function of QUE on the Th1/Th2 immune imbalance that is induced by MG infection in the tracheal mucosa. Moreover, QUE intervention impeded the M1 polarization of HD-11 cells induced by MG infection, while simultaneously promoting M2 polarization through the induction of FAO. Conversely, inhibitors of the FAO pathway impede this effect. The results of computer network analysis suggest that QUE may induce FAO via the PI3K/AKT pathway to promote M2 polarization. Notably, inhibition of the PI3K/AKT pathway was found to effectively inhibit M2 polarization in HD-11 cells, while having a limited effect on FAO. CONCLUSIONS: QUE promotes M2 polarization of HD-11 cells to enhance Th2 immune response through FAO and PI3K/AKT pathways, thereby restoring tracheal mucosal barrier function and ultimately inhibiting MG colonization.


Subject(s)
Chickens , Mycoplasma Infections , Mycoplasma gallisepticum , Poultry Diseases , Quercetin , Th2 Cells , Animals , Quercetin/pharmacology , Mycoplasma gallisepticum/drug effects , Mycoplasma Infections/drug therapy , Mycoplasma Infections/immunology , Poultry Diseases/drug therapy , Poultry Diseases/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Respiratory Mucosa/drug effects , Respiratory Mucosa/immunology , Trachea/drug effects , Molecular Docking Simulation , Tight Junctions/drug effects , Immunoglobulin A, Secretory/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Th1 Cells/drug effects , Th1 Cells/immunology , Macrophages/drug effects , Fatty Acids
20.
JCI Insight ; 9(14)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-39133648

ABSTRACT

Neutrophils (polymorphonuclear leukocytes, PMNs) comprise a major component of the immune cell infiltrate during acute mucosal inflammation and have an important role in molding the inflammatory tissue environment. While PMNs are essential to clearance of invading microbes, the major PMN antimicrobial enzyme myeloperoxidase (MPO) can also promote bystander tissue damage. We hypothesized that blocking MPO would attenuate acute colitis and prevent the development of chronic colitis by limiting bystander tissue damage. Using the acute and chronic dextran sodium sulfate model of murine colitis, we demonstrated that MPO-deficient mice experienced less inflammation and more rapidly resolved colitis relative to wild-type controls. Mechanistic studies demonstrated that activated MPO disrupted intestinal epithelial barrier function through the dysregulation of the epithelial tight junction proteins. Our findings revealed that activated MPO chlorinates tyrosine within several tight junction proteins, thereby promoting tight junction mislocalization and dysfunction. These observations in cell models and in murine colitis were validated in human intestinal biopsies from individuals with ulcerative colitis and revealed a strong correlation between disease severity (Mayo score) and tissue chlorinated tyrosine levels. In summary, these findings implicate MPO as a viable therapeutic target to limit bystander tissue damage and preserve mucosal barrier function during inflammation.


Subject(s)
Disease Models, Animal , Intestinal Mucosa , Neutrophils , Peroxidase , Tight Junction Proteins , Peroxidase/metabolism , Animals , Mice , Humans , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Neutrophils/metabolism , Neutrophils/immunology , Tight Junction Proteins/metabolism , Colitis/pathology , Colitis/metabolism , Colitis/chemically induced , Halogenation , Inflammation/metabolism , Inflammation/pathology , Male , Mice, Knockout , Dextran Sulfate/toxicity , Tight Junctions/metabolism , Female , Mice, Inbred C57BL , Colitis, Ulcerative/pathology , Colitis, Ulcerative/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL