Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 220
Filter
1.
Front Immunol ; 13: 955576, 2022.
Article in English | MEDLINE | ID: mdl-36091010

ABSTRACT

The uterine endometrium uniquely regenerates after menses, postpartum, or after breaks in the uterine layer integrity throughout women's lives. Direct cell-cell contacts ensured by tight and adherens junctions play an important role in endometrial integrity. Any changes in these junctions can alter the endometrial permeability of the uterus and have an impact on the regeneration of uterine layers. Interleukin 22 (IL-22) is a cytokine that is recognized for its role in epithelial regeneration. Moreover, it is crucial in controlling the inflammatory response in mucosal tissues. Here, we studied the role of IL-22 in endometrial recovery after inflammation-triggered abortion. Fecundity of mice was studied in consecutive matings of the same animals after lipopolysaccharide (LPS) (10 µg per mouse)-triggered abortion. The fecundity rate after the second mating was substantially different between IL-22 knockout (IL-22-/-) (9.1%) and wild-type (WT) (71.4%) mice (p < 0.05), while there was no difference between the groups in the initial mating, suggesting that IL-22 deficiency might be associated with secondary infertility. A considerable difference was observed between IL-22-/- and WT mice in the uterine clearance following LPS-triggered abortion. Gross examination of the uteri of IL-22-/- mice revealed non-viable fetuses retained inside the horns (delayed clearance). In contrast, all WT mice had completed abortion with total clearance after LPS exposure. We also discovered that IL-22 deficiency is associated with a decreased expression of tight junctions (claudin-2 and claudin-10) and cell surface pathogen protectors (mucin-1). Moreover, IL-22 has a role in the remodeling of the uterine tissue in the inflammatory environment by regulating epithelial-mesenchymal transition markers called E- and N-cadherin. Therefore, IL-22 contributes to the proper regeneration of endometrial layers after inflammation-triggered abortion. Thus, it might have a practical significance to be utilized as a treatment option postpartum (enhanced regeneration function) and in secondary infertility caused by inflammation (enhanced barrier/protector function).


Subject(s)
Endometrium , Extracellular Matrix , Inflammation , Interleukins , Regeneration , Tight Junctions , Abortion, Spontaneous/immunology , Animals , Endometrium/immunology , Extracellular Matrix/genetics , Extracellular Matrix/immunology , Female , Humans , Infertility/genetics , Infertility/immunology , Inflammation/genetics , Inflammation/immunology , Interleukins/genetics , Interleukins/immunology , Lipopolysaccharides/immunology , Mice , Pregnancy , Regeneration/immunology , Tight Junctions/immunology , Interleukin-22
2.
Nat Commun ; 13(1): 874, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35169117

ABSTRACT

IL-18 is emerging as an IL-22-induced and epithelium-derived cytokine which contributes to host defence against intestinal infection and inflammation. In contrast to its known role in Goblet cells, regulation of barrier function at the molecular level by IL-18 is much less explored. Here we show that IL-18 is a bona fide IL-22-regulated gate keeper for intestinal epithelial barrier. IL-22 promotes crypt immunity both via induction of phospho-Stat3 binding to the Il-18 gene promoter and via Il-18 independent mechanisms. In organoid culture, while IL-22 primarily increases organoid size and inhibits expression of stem cell genes, IL-18 preferentially promotes organoid budding and induces signature genes of Lgr5+ stem cells via Akt-Tcf4 signalling. During adherent-invasive E. coli (AIEC) infection, systemic administration of IL-18 corrects compromised T-cell IFNγ production and restores Lysozyme+ Paneth cells in Il-22-/- mice, but IL-22 administration fails to restore these parameters in Il-18-/- mice, thereby placing IL-22-Stat3 signalling upstream of the IL-18-mediated barrier defence function. IL-18 in return regulates Stat3-mediated anti-microbial response in Paneth cells, Akt-Tcf4-triggered expansion of Lgr5+ stem cells to facilitate tissue repair, and AIEC clearance by promoting IFNγ+ T cells.


Subject(s)
Escherichia coli Infections/immunology , Immunity, Mucosal/immunology , Interleukin-18/immunology , Interleukins/immunology , Intestinal Mucosa/immunology , Animals , Crohn Disease/microbiology , Crohn Disease/pathology , Dysbiosis/microbiology , Escherichia coli/immunology , Interferon-gamma/immunology , Interleukin-18/genetics , Intestinal Mucosa/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muramidase/metabolism , Organoids , Paneth Cells/immunology , Promoter Regions, Genetic/genetics , STAT3 Transcription Factor/metabolism , Tight Junctions/immunology , Interleukin-22
3.
J Immunol ; 208(2): 227-234, 2022 01 15.
Article in English | MEDLINE | ID: mdl-35017212

ABSTRACT

Our body's most outward facing epithelial barrier, the skin, serves as the frontline defense against myriad environmental assailants. To combat these motley threats, the skin has evolved a sophisticated immunological arsenal. In this article, I provide an overview of the skin's complex architecture and the distinct microniches in which immune cells reside and function. I review burgeoning literature on the synchronized immune, stromal, epithelial, and neuronal cell responses in healthy and inflamed skin. Next, I delve into the distinct requirement and mechanisms of long-term immune surveillance and tissue adaptation at the cutaneous frontier. Finally, by discussing the contributions of immune cells in maintaining and restoring tissue integrity, I underscore the constellation of noncanonical functions undertaken by the skin immune system. Just as our skin's immune system benefits from embracing diverse defense strategies, so, too, must we in the immunology research community support disparate perspectives and people from all walks of life.


Subject(s)
Immune System Phenomena/physiology , Immunologic Surveillance/immunology , Skin/immunology , Humans , Immune System/immunology , Skin/anatomy & histology , Tight Junctions/immunology
4.
J Sci Food Agric ; 102(1): 434-444, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34143895

ABSTRACT

BACKGROUND: The incidence of inflammatory bowel disease (IBD) continues to increase worldwide. Multiple factors, including diet, loss of the intestinal barrier function, and imbalance of the immune system can cause IBD. A balanced diet is important for maintaining a healthy bowel and preventing IBD from occurring. The effects of probiotic Lactobacillus gasseri-fermented Maillard reaction products (MRPs) prepared by reacting whey protein with galactose on anti-inflammation and intestinal homeostasis were investigated in this study, which compared MPRs and probiotics separately. RESULTS: In an animal colitis model induced by 2% dextran sulfate sodium (DSS), FWG administration alleviated colon length loss and maintained intestinal immune system homeostasis as reflected by down-regulated interleukin (IL)-6, IL-10, tumor necrosis factor (TNF)-α output, and metallopeptidase-9, and epithelial barrier balance as reflected by up-regulated occludin, E-cadherin, and zonula occludens-1 production in the colon. Furthermore, the expression of splenic cytokines such as IL-6, TNF-α, and IL-10 was up-regulated in the FWG-treated mice in a comparable amount to the control group to ensure the balance of immune responses. CONCLUSION: This study showed that the use of FWG protects the intestines from colitis caused by DSS and maintains immune balance. FWG increased antioxidant enzyme activity, increased intestinal permeability, and regulated the balance of pro- and anti-inflammatory cytokines in the intestines and spleen. Continued intake of FWG can alleviate IBD symptoms through the preservation of mucosal immune responses, epithelial junction and homeostasis through the regulated splenic cytokines. © 2021 Society of Chemical Industry.


Subject(s)
Colitis/drug therapy , Glycation End Products, Advanced/administration & dosage , Lactobacillus gasseri/metabolism , Probiotics/administration & dosage , Animals , Anti-Inflammatory Agents/administration & dosage , Colitis/chemically induced , Colitis/immunology , Colitis/physiopathology , Colon/drug effects , Colon/immunology , Dextran Sulfate/adverse effects , Disease Models, Animal , Galactose/metabolism , Glycation End Products, Advanced/metabolism , Homeostasis/drug effects , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Male , Mice , Mice, Inbred C57BL , Tight Junctions/genetics , Tight Junctions/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Whey Proteins/metabolism
5.
Front Immunol ; 12: 767505, 2021.
Article in English | MEDLINE | ID: mdl-34712246

ABSTRACT

Interferon λ (IFN-λ) is critical for host viral defense at mucosal surfaces and stimulates immunomodulatory signals, acting on epithelial cells and few other cell types due to restricted IFN-λ receptor expression. Epithelial cells of the intestine play a critical role in the pathogenesis of Inflammatory Bowel Disease (IBD), and the related type II interferons (IFN-γ) have been extensively studied in the context of IBD. However, a role for IFN-λ in IBD onset and progression remains unclear. Recent investigations of IFN-λ in IBD are beginning to uncover complex and sometimes opposing actions, including pro-healing roles in colonic epithelial tissues and potentiation of epithelial cell death in the small intestine. Additionally, IFN-λ has been shown to act through non-epithelial cell types, such as neutrophils, to protect against excessive inflammation. In most cases IFN-λ demonstrates an ability to coordinate the host antiviral response without inducing collateral hyperinflammation, suggesting that IFN-λ signaling pathways could be a therapeutic target in IBD. This mini review discusses existing data on the role of IFN-λ in the pathogenesis of inflammatory bowel disease, current gaps in the research, and therapeutic potential of modulating the IFN-λ-stimulated response.


Subject(s)
Epithelial Cells/immunology , Immunity, Innate/immunology , Inflammatory Bowel Diseases/immunology , Interferons/immunology , Intestinal Mucosa/immunology , Signal Transduction/immunology , Animals , Apoptosis/immunology , Cytokines/immunology , Cytokines/metabolism , Epithelial Cells/metabolism , Humans , Inflammatory Bowel Diseases/metabolism , Interferons/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Models, Immunological , Protein Isoforms/immunology , Protein Isoforms/metabolism , STAT Transcription Factors/immunology , STAT Transcription Factors/metabolism , Tight Junctions/immunology , Tight Junctions/metabolism , Interferon Lambda
6.
Int Arch Allergy Immunol ; 182(11): 1046-1057, 2021.
Article in English | MEDLINE | ID: mdl-34148047

ABSTRACT

BACKGROUND: Tumor protein p63 has been shown to be important for epithelial dysfunction, including epithelial barrier defects and mucosal inflammation, in the development of chronic rhinosinusitis with nasal polyps (CRSwNP). Basonuclin1 (BNC1), an epithelial-specific transcriptional factor, is a direct downstream target of p63 and thus might be involved in the pathogenesis of CRSwNP. OBJECTIVE: We sought to investigate whether BNC1 was associated with p63-mediated epithelial barrier defects and nasal mucosal inflammation in CRSwNP. METHODS: Nasal tissue biopsies were obtained from 91 patients to CRSwNP, 49 chronic rhinosinusitis without nasal polyps (CRSsNP) patients, and 28 control subjects. Immunohistochemistry and immunofluorescence staining were used to determine the distribution of BNC1 in tissues and localization in cells, respectively. Quantitative PCR was performed to detect the expression levels of BNC1, TP63, epithelial barrier proteins, and type-2 helper T-cell inflammation-related genes. RESULTS: BNC1 mRNA expression was significantly elevated in the tissues in CRSwNP patients compared with CRSsNP (1.96-fold, p = 0.0003) and control groups (2.40-fold, p < 0.0001). BNC1 staining was strongly positive in the nasal epithelium and co-localized with p63-positive epithelial cells. The expression of BNC1 mRNA was strongly correlated with TP63 mRNA level both in tissue biopsies (r = 0.78, p < 0.0001) and epithelial scrapings (r = 0.97, p < 0.0001). BNC1 expression was also positively correlated with epithelial barrier protein genes (CDH1, CLDN1, CLDN4, TJP1, and TJP2) and epithelial genes involved in TH2 inflammation (IL33, CCL26, CLC, and ALOX15). CONCLUSIONS: Overexpression of BNC1 may be associated with increased expression of TP63, and possibly contribute to the epithelial barrier defects and TH2 inflammation in CRSwNP.


Subject(s)
DNA-Binding Proteins/genetics , Nasal Mucosa/immunology , Nasal Polyps/immunology , Rhinitis/immunology , Sinusitis/immunology , Th2 Cells/immunology , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Adult , Chronic Disease , DNA-Binding Proteins/immunology , Female , Humans , Male , Middle Aged , Tight Junction Proteins/genetics , Tight Junction Proteins/immunology , Tight Junctions/immunology , Transcription Factors/immunology , Tumor Suppressor Proteins/immunology , Up-Regulation
7.
Front Immunol ; 12: 637603, 2021.
Article in English | MEDLINE | ID: mdl-33717185

ABSTRACT

The role of dietary components in immune function has acquired considerable attention in recent years. An important focus area is to unravel the role of bioactive dietary compounds in relation to enteric disease and their impact on gut mucosal immunity. Proanthocyanidins (PAC) are among the most common and most consumed dietary polyphenols, and are characterised by their variable molecular structures and diverse bioactivities. In particular, their anti-oxidative effects and ability to modulate gut microbiota have been widely described. However, there is limited evidence on the mechanism of action of PAC on the immune system, nor is it clearly established how PAC may influence susceptibility to enteric infections. Establishing the sites of action of PAC and their metabolites within the gut environment is fundamental to determine the applicability of PAC against enteric pathogens. Some mechanistic studies have shown that PAC have direct modulatory effects on immune cell signalling, isolated pathogens, and gut mucosal barrier integrity. Boosting the recruitment of immune cells and suppressing the amount of pro-inflammatory cytokines are modulating factors regulated by PAC, and can either be beneficial or detrimental in the course of re-establishing gut homeostasis. Herein, we review how PAC may alter distinct immune responses towards enteric bacterial, viral and parasitic infections, and how the modulation of gut microbiota may act as a mediating factor. Furthermore, we discuss how future studies could help unravel the role of PAC in preventing and/or alleviating intestinal inflammation and dysbiosis caused by enteric disease.


Subject(s)
Antioxidants/pharmacology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Proanthocyanidins/pharmacology , Tight Junctions/physiology , Antioxidants/administration & dosage , Diet , Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Humans , Immunity, Mucosal/immunology , Proanthocyanidins/administration & dosage , Tight Junctions/immunology , Tight Junctions/microbiology
8.
Physiol Rep ; 9(3): e14717, 2021 02.
Article in English | MEDLINE | ID: mdl-33523589

ABSTRACT

Piglets must acquire passive immunity through colostrum within hours after birth to survive. How colostral macromolecules traverse the small intestinal epithelium may include nonselective pinocytosis and paracellular transport through tight junction proteins located between epithelial cells. Claudin proteins-3 and -4 contribute to the epithelial tight junctions (TJs) on the apical aspect of lateral surfaces of intestinal epithelial cells (IECs) where they help regulate ion and macromolecule movement across the intestinal epithelium. Throughout the small intestine of newborn piglets, Claudin-3 was localized to the lateral and basolateral surface of intestinal epithelial cells as well as the membrane of large vacuoles. In the duodenum and jejunum, Claudin-4 was localized to the apical surface independent of tight junction regions. In the ileum, Claudin-4 was localized to the lateral and basolateral surfaces indicating region-specific differences and noncanonical patterns of Claudin-4 localization independent of tight junction regions. Understanding the timing of changes in surface localization of Claudin-3 and Claudin-4 and how they may coincide with changes in small intestinal permeability may help develop new protective strategies against infectious diseases within newborn piglets.


Subject(s)
Claudin-3/metabolism , Claudin-4/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Tight Junctions/metabolism , Animals , Animals, Newborn , Biological Transport , Colostrum/metabolism , Immunity , Intestinal Mucosa/immunology , Intestine, Small/immunology , Permeability , Sus scrofa , Tight Junctions/immunology
9.
Int J Mol Sci ; 22(3)2021 Jan 30.
Article in English | MEDLINE | ID: mdl-33573189

ABSTRACT

Hyperglycemia alters the function of cerebral endothelial cells from the blood-brain barrier, increasing the risk of cerebrovascular complications during diabetes. This study evaluated the protective effect of polyphenols on inflammatory and permeability markers on bEnd3 cerebral endothelial cells exposed to high glucose concentration. Results show that hyperglycemic condition increased nuclear factor kappa B (NFκB) activity, deregulated the expression of interleukin-1 beta (IL-1ß), interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), interleukin-10 (IL-10) and endothelial-leukocyte adhesion molecule (E-selectin) genes, raised MCP-1 secretion and elevated monocyte adhesion and transendothelial migration. High glucose decreased occludin, claudin-5, zona occludens-1 (ZO-1) and zona occludens-2 (ZO-2) tight junctions production and altered the endothelial permeability. Characterized polyphenolic extracts from the French medicinal plants Antirhea borbonica, Ayapana triplinervis, Dodonaea viscosa and Terminalia bentzoe, and their major polyphenols quercetin, caffeic, chlorogenic and gallic acids limited the pro-inflammatory and permeability alterations caused by high glucose. Peroxisome proliferator-activated receptor gamma (PPARγ) agonist also attenuated these damages while PPARγ antagonist aggravated them, suggesting PPARγ protective action. Interestingly, polyphenols improved PPARγ gene expression lowered by high glucose. Moreover, polyphenols were detected at the intracellular level or membrane-bound to cells, with evidence for breast cancer resistance protein (BCRP) efflux transporter role. Altogether, these findings emphasize the ability of polyphenols to protect cerebral endothelial cells in hyperglycemic condition and their relevance for pharmacological strategies aiming to limit cerebrovascular disorders in diabetes.


Subject(s)
Blood-Brain Barrier/drug effects , Cerebrovascular Disorders/prevention & control , Hyperglycemia/immunology , Plant Extracts/pharmacology , Polyphenols/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Blood Glucose/metabolism , Blood-Brain Barrier/cytology , Blood-Brain Barrier/immunology , Blood-Brain Barrier/pathology , Capillary Permeability/drug effects , Capillary Permeability/immunology , Cell Line , Cerebrovascular Disorders/etiology , Cerebrovascular Disorders/immunology , Cerebrovascular Disorders/pathology , Drug Evaluation, Preclinical , Endothelial Cells/immunology , Endothelial Cells/pathology , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Humans , Hyperglycemia/blood , Hyperglycemia/complications , Mice , Monocytes/drug effects , Monocytes/immunology , NF-kappa B/metabolism , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Plant Extracts/therapeutic use , Polyphenols/therapeutic use , Signal Transduction/drug effects , Tight Junctions/drug effects , Tight Junctions/immunology , Tight Junctions/pathology
10.
Biomed Pharmacother ; 137: 111270, 2021 May.
Article in English | MEDLINE | ID: mdl-33485121

ABSTRACT

Our previous studies have shown that the environmental contaminant bisphenol A (BPA) exhibits strong intestinal toxicity and can readily cause intestinal barrier dysfunction. However, the causal relationship between adverse biological processes of BPA-induced intestinal tissue and the role of key signaling molecules in it requires further investigation. In this study, we established a mouse and intestinal epithelial cell model of BPA treatment to determine the underlying molecular mechanisms of BPA-induced intestinal injury. The results showed that the BPA treatment increased the intestinal permeability and disrupted the barrier function by increasing the chemical marker content and tight junction expression in intestinal tissues and blood circulation. BPA also altered the oxidative and antioxidant status of intestinal epithelial cells by increasing ROS and RNS contents and decreasing the activity levels of SOD, GPx, CAT, and T-AOC. BPA further induced inflammatory responses by upregulating the gene abundance of key factors of the innate immune system (TLR2, TLR4, MyD88, and NF-κB), the transcriptional activity of NF-kB, and the secretion of pro-inflammatory cytokines (IL-1ß, IL-6, IL-8, and TNF-α). Moreover, apoptosis was activated by BPA, whereas cell proliferation was inhibited by BPA. Mechanistically, co-treatment of intestinal epithelial cells with BPA using the oxidative stress scavenger NAC, the NF-κB-specific inhibitor JSH-23, and the apoptosis inhibitor Z-VAD-FMK, respectively, showed that BPA activates the innate immune response by inducing oxidative stress. Consequently, apoptosis is promoted, and cell proliferation is inhibited, ultimately disrupting the intestinal barrier function. Our findings provide insight into the pathogenesis of BPA-induced gut injury.


Subject(s)
Benzhydryl Compounds/toxicity , Colon/drug effects , Environmental Pollutants/toxicity , Intestinal Mucosa/drug effects , Phenols/toxicity , Tight Junctions/drug effects , Animals , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Colon/immunology , Colon/metabolism , Colon/pathology , Cytokines/metabolism , Immunity, Innate/drug effects , Inflammation Mediators/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , Oxidative Stress/drug effects , Permeability , Reactive Oxygen Species/metabolism , Signal Transduction , Tight Junctions/immunology , Tight Junctions/metabolism , Tight Junctions/pathology
11.
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
12.
Food Chem Toxicol ; 145: 111712, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32877744

ABSTRACT

Lipopolysaccharide (LPS) is the key factor in various intestinal inflammation which could disrupt the epithelial barrier function. Deoxynivalenol (DON), a well-known mycotoxin, can induce intestinal injury. However, the combined enterotoxicity of LPS and DON has rarely been studied. In this study, IPEC-J2 cell monolayers were exposed to LPS and nontoxic-dose DON for 12 and 24 h to investigate the effects of DON on LPS-induced inflammatory response and tight junction variation, and specific inhibitor and CRISPR-Cas9 were used to explore the underlying mechanisms. Our results showed that nontoxic-dose DON aggravated LPS-induced cellular inflammatory response, reflecting on more significant changes of inflammatory cytokines mRNA expression, higher protein expression of NOD-like receptor protein 3 (NLRP3) and procaspase-1. Moreover, nontoxic-dose DON aggravated LPS-induced mRNA and protein expression decreased, and distribution confused of tight junction proteins. We found that DON further enhanced LPS-induced phosphorylation and nucleus translocation of p65, and expression of LC3B-Ⅱ. NF-κB inhibitor and CRISPR-Cas9-mediated knockout of LC3B attenuated the effects of combination which indicated nontoxic-dose DON aggravated LPS-induced intestinal inflammation and tight junction disorder through activating NF-κB signaling pathway and autophagy-related protein LC3B. It further warns that ingesting low doses of mycotoxins may exacerbate the effects of intestinal pathogens on the body.


Subject(s)
Inflammation/immunology , Microtubule-Associated Proteins/immunology , NF-kappa B/immunology , Tight Junctions/drug effects , Trichothecenes/toxicity , Animals , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/immunology , Inflammation/etiology , Inflammation/genetics , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Lipopolysaccharides/adverse effects , Microtubule-Associated Proteins/genetics , NF-kappa B/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Signal Transduction/drug effects , Tight Junctions/immunology
13.
Food Funct ; 11(9): 8193-8201, 2020 Sep 23.
Article in English | MEDLINE | ID: mdl-32966466

ABSTRACT

Long-term intake of a high-fat diet seriously affects the health of pregnant women and leads to increased levels of inflammation in the mammary gland. Therefore, to further explore the effect of a high-fat diet on mammary gland development and the tight junction (TJ) during pregnancy, we placed mice into two groups: a high-fat diet group and a control group. We detected the expression of proteins related to fat synthesis in the mammary gland by western blotting. The results showed that a high-fat diet could lead to an increase in fat synthesis in the mammary gland. Then, the inflammatory levels and acinar cell morphology in the mammary gland were detected by ELISA and H&E staining. We also measured the levels of MAPK and NF-κB signal pathway-related proteins by western blotting. The results showed that a high-fat diet activated the MAPK and NF-κB signaling pathways and promoted the expression of inflammatory factors. Finally, the development of the mammary gland and the integrity of the TJ were determined by immunohistochemistry, immunofluorescence and western blotting. The results showed that a high-fat diet inhibited the development of the mammary gland and the expression of tight junction proteins (TJs). Our study showed that a high-fat diet could promote the expression of inflammatory factors by activating the MAPK and NF-κB signaling pathways and could reshape the microenvironment through extramammary inflammation. Finally, a high-fat diet inhibited the development of the mammary gland during pregnancy and destroyed the integrity of the TJ.


Subject(s)
Diet, High-Fat/adverse effects , Mammary Glands, Human/growth & development , Pregnancy/immunology , Tight Junctions/immunology , Animals , Female , Humans , Male , Mammary Glands, Human/immunology , Mice , Mice, Inbred ICR , NF-kappa B/genetics , NF-kappa B/immunology , Pregnancy/genetics , Signal Transduction , Tight Junction Proteins/genetics , Tight Junction Proteins/immunology
14.
Article in English | MEDLINE | ID: mdl-32637365

ABSTRACT

The airway epithelial barrier is a major barrier protecting against clinically significant infections of the lung. Its integrity is often compromised due to mechanical, chemical, or infectious causes. Opportunistic bacterial pathogens are poised to cause parenchymal infection and become difficult to eradicate due to adaptive metabolic changes, biofilm formation, and the acquisition of antimicrobial resistance and fitness genes. Enhancing mucosal defenses by modulating the cytokines that regulate barrier functions, such as interleukin-22 (IL-22) and interferon-λ (IFN-λ), members of the IL-10 family of cytokines, is an attractive approach to prevent these infections that are associated with high morbidity and mortality. These cytokines both signal through the cognate receptor IL-10RB, have related protein structures and common downstream signaling suggesting shared roles in host respiratory defense. They are typically co-expressed in multiple models of infections, but with differing kinetics. IL-22 has an important role in the producing antimicrobial peptides, upregulating expression of junctional proteins in the airway epithelium and working in concert with other inflammatory cytokines such as IL-17. Conversely, IFN-λ, a potent antiviral in influenza infection with pro-inflammatory properties, appears to decrease junctional integrity allowing for bacterial and immune cell translocation. The effects of these cytokines are pleotropic, with pathogen and tissue specific consequences. Understanding how these cytokines work in the mucosal defenses of the respiratory system may suggest potential targets to prevent invasive infections of the damaged lung.


Subject(s)
Interferon-gamma/immunology , Interleukin-10 Receptor beta Subunit/immunology , Interleukins/immunology , Respiratory Mucosa/immunology , Tight Junctions/immunology , Coronavirus Infections/immunology , Humans , Influenza, Human/immunology , Klebsiella Infections/immunology , Klebsiella pneumoniae/immunology , Pseudomonas aeruginosa/immunology , Respiratory Mucosa/microbiology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Interleukin-22
15.
PLoS Negl Trop Dis ; 14(6): e0008335, 2020 06.
Article in English | MEDLINE | ID: mdl-32511241

ABSTRACT

Zika virus (ZIKV), a neglected tropical disease until its re-emergence in 2007, causes microcephaly in infants and Guillain-Barré syndrome in adults. Its re-emergence and spread to more than 80 countries led the World Health Organization in 2016 to declare a Public Health Emergency. ZIKV is mainly transmitted by mosquitos, but can persist in infected human male semen for prolonged periods and may be sexually transmitted. Testicular Sertoli cells support ZIKV replication and may be a reservoir for persistent ZIKV infection. Electrical impedance analyses indicated ZIKV infection rapidly disrupted Vero cell monolayers but had little effect upon human Sertoli cells (HSerC). We determined ZIKV-induced proteomic changes in HSerC using an aptamer-based multiplexed technique (SOMAscan) targeting >1300 human proteins. ZIKV infection caused differential expression of 299 proteins during three different time points, including 5 days after infection. Dysregulated proteins are involved in different bio-functions, including cell death and survival, cell cycle, maintenance of cellular function, cell signaling, cellular assembly, morphology, movement, molecular transport, and immune response. Many signaling pathways important for maintenance of HSerC function and spermatogenesis were highly dysregulated. These included IL-6, IGF1, EGF, NF-κB, PPAR, ERK/MAPK, and growth hormone signaling. Down-regulation of the PPAR signaling pathway might impact cellular energy supplies. Upstream molecule analysis also indicated microRNAs involved in germ cell development were downregulated by infection. Overall, this study leads to a better understanding of Sertoli cellular mechanisms used by ZIKV during persistent infection and possible ZIKV impacts on spermatogenesis.


Subject(s)
Sertoli Cells/immunology , Spermatogenesis , Tight Junctions/immunology , Zika Virus Infection/immunology , Animals , Chlorocebus aethiops , Humans , Male , Proteomics , Semen/virology , Sertoli Cells/virology , Signal Transduction , Tight Junctions/virology , Vero Cells , Virus Replication , Zika Virus
16.
J Leukoc Biol ; 108(6): 1735-1746, 2020 12.
Article in English | MEDLINE | ID: mdl-32573820

ABSTRACT

Recent studies have revealed that aloe emodin (AE), a natural compound from the root and rhizome of Rheum palmatum L., exhibits significant pharmacologic activities. However, the pharmacologic relevance of the compound, particularly for cardiovascular disease, remains largely unknown. Here, we hypothesized that AE could improve endothelial junction dysfunction through inhibiting the activation of NOD-like receptor family pyrin domain containing-3 (NLRP3) inflammasome regulated by NLRP3 ubiquitination, and ultimately prevent cardiovascular disease. In vivo, we used confocal microscopy to study the expression of tight junction proteins zonula occludens-1/2 (ZO-1/2) and the formation of NLRP3 inflammasome in coronary arteries of hypertension. And the experimental serum was used to detect the activation of NLRP3 inflammasome by ELISA assay. We found that AE could restore the expression of the endothelial connective proteins ZO-1/2 and decrease the release of high mobility group box1 (HMGB1), and also inhibited the formation and activation of NLRP3 inflammasome. Similarly, in vitro, our findings demonstrated that AE could restore the expression of the tight junction proteins ZO-1/2 and decrease monolayer cell permeability that related to endothelial function after stimulation by angiotensin II (Ang II) in microvascular endothelial cells (MECs). We also demonstrated that AE could inhibit Ang II-induced NLRP3 inflammasome formation and activation, which were regulated by NLRP3 ubiquitination in MECs, as shown by fluorescence confocal microscopy and Western blot. Together with these changes, we revealed a new protection mechanism of AE that inhibited NLRP3 inflammasome activation and decreased the release of HMGB1 by promoting NLRP3 ubiquitination. Our findings implicated that AE exhibited immense potential and specific therapeutic value in hypertension-related cardiovascular disease in the early stage and the development of innovative drugs.


Subject(s)
Angiotensin II/adverse effects , Anthraquinones/pharmacology , Endothelial Cells/immunology , Inflammasomes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Tight Junctions/immunology , Ubiquitination/drug effects , Angiotensin II/pharmacology , Animals , Endothelial Cells/pathology , HMGB1 Protein/immunology , Male , Mice , Tight Junctions/pathology , Ubiquitination/immunology , Zonula Occludens-1 Protein/immunology , Zonula Occludens-2 Protein/immunology
17.
Toxicol Lett ; 331: 208-217, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32569800

ABSTRACT

Fine particulate matter 2.5 (PM2.5), one of the main components of air pollutants, seriously threatens human health. Possible neuronal dysfunction induced by PM2.5 has received extensive attention. However, there is little evidence for the specific biochemical mechanism of neuronal injury induced by PM2.5. Moreover, the pathway for PM2.5 transport from peripheral circulation to the central nervous system (CNS) is still unclear. In the current work, C57BL/6 mice were chronically exposed to ambient PM2.5 for 3, 6, 9, and 12 months. Exposure to ambient PM2.5 resulted in a significant reduction of cognitive ability in mice by Morris water maze test. PM2.5 exposure induced a neuroinflammatory reaction after cognitive impairment, while inflammation in the hypothalamus and olfactory bulb tissue occurred earlier. The expression levels of integrity tight junction proteins in the blood-brain barrier (BBB) were reduced by PM2.5 exposure. Pulmonary inflammation occurred much earlier and diminished at later stage of PM2.5 exposure. The results indicated that chronic exposure to ambient PM2.5 led to cognitive decline in mice; CNS dysfunction may be due to neuroinflammatory reactions; the reduced integrity of the BBB allowed the influence of pulmonary inflammation to neuronal alterations. The work may provide promising therapeutic or preventive targets for air pollution-induced neurodegenerative disease.


Subject(s)
Air Pollutants/toxicity , Cognitive Dysfunction/chemically induced , Inhalation Exposure/adverse effects , Neurodegenerative Diseases/chemically induced , Particulate Matter/toxicity , Pneumonia/chemically induced , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/immunology , Cognitive Dysfunction/immunology , Cytokines/metabolism , Dose-Response Relationship, Drug , Male , Maze Learning/drug effects , Mice, Inbred C57BL , Neurodegenerative Diseases/immunology , Particle Size , Pneumonia/immunology , Tight Junctions/drug effects , Tight Junctions/immunology , Up-Regulation
18.
Fish Shellfish Immunol ; 103: 211-219, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32422190

ABSTRACT

The current study was designed to evaluate the effect of Yucca schidigera extract (YSE) on the growth performance, intestinal antioxidant status, immune response, and tight junctions of mirror carp (Cyprinus carpio). A total of 450 mirror carp (45.21 ± 0.43 g) were fed diets supplemented with 0, 200, or 400 mg/kg YSE for 8 weeks. Compared with the control (0 mg/kg), the final body weight and weight gain rate were significantly higher in the 400 mg/kg YSE group (P < 0.05), and the serum ammonia concentration was significantly lower in both YSE groups (P < 0.05). Additionally, the total antioxidant capacity was significantly higher in the 400 mg/kg YSE group (P < 0.05), and the malondialdehyde content was significantly lower in both YSE groups (P < 0.05). Complement 3 and 4 contents were significantly higher in the 400 mg/kg YSE group (P < 0.05), and lysozyme was significantly higher in both YSE groups compared to the control group (P < 0.05). The relative mRNA levels of copper zinc superoxide dismutase, catalase, glutathione peroxidase1a, and nuclear factor erythroid 2-related factor 2 as well as transforming growth factor ß were significantly higher in both YSE supplemented groups compared to the control (P < 0.05), whereas the relative mRNA level of Kelch-like ECH-associated protein 1 was significantly lower in both YSE supplemented groups (P < 0.05). The relative mRNA levels of interleukin 1ß and interleukin 6 were significantly lower in the 400 mg/kg YSE supplemented group compared to the control (P < 0.05). Additionally, both YSE levels decreased the relative mRNA expression of tumour necrosis factor-α (P < 0.05). The relative mRNA levels of ZO-1 and claudin 11 were significantly higher in both YSE supplemented groups (P < 0.05), and the relative mRNA level of occludin was significantly higher in the 200 mg/kg YSE group than the control and 400 mg/kg YSE groups (P < 0.05). In conclusion, dietary supplementation with 400 mg/kg YSE improved the growth, intestinal antioxidant status, immune response, and tight junctions of mirror carp.


Subject(s)
Antioxidants/metabolism , Carps/immunology , Immunity , Intestines/immunology , Plant Extracts/metabolism , Tight Junctions/immunology , Yucca/chemistry , Animal Feed/analysis , Animals , Carps/growth & development , Carps/metabolism , Diet/veterinary , Dietary Supplements/analysis , Dose-Response Relationship, Drug , Immunity/drug effects , Intestines/drug effects , Plant Extracts/administration & dosage , Random Allocation , Tight Junctions/drug effects
19.
J Allergy Clin Immunol ; 145(6): 1641-1654, 2020 06.
Article in English | MEDLINE | ID: mdl-32305348

ABSTRACT

BACKGROUND: Prenatal challenges such as maternal stress perception increase the risk and severity of asthma during childhood. However, insights into the trajectories and targets underlying the pathogenesis of prenatally triggered asthma are largely unknown. The developing lung and immune system may constitute such targets. OBJECTIVE: Here we have aimed to identify the differential sex-specific effects of prenatal challenges on lung function, immune response, and asthma severity in mice. METHODS: We generated bone marrow chimeric (BMC) mice harboring either prenatally stress-exposed lungs or a prenatally stress-exposed immune (hematopoietic) system and induced allergic asthma via ovalbumin. Next-generation sequencing (RNA sequencing) of lungs and assessment of airway epithelial barrier function in ovalbumin-sensitized control and prenatally stressed offspring was also performed. RESULTS: Profoundly enhanced airway hyperresponsiveness, inflammation, and fibrosis were exclusively present in female BMC mice with prenatally stress-exposed lungs. These effects were significantly perpetuated if both the lungs and the immune system had been exposed to prenatal stress. A prenatally stress-exposed immune system alone did not suffice to increase the severity of these asthma features. RNA sequencing analysis of lungs from prenatally stressed, non-BMC, ovalbumin-sensitized females unveiled a deregulated expression of genes involved in asthma pathogenesis, tissue remodeling, and tight junction formation. It was also possible to independently confirm a tight junction disruption. In line with this, we identified an altered perinatal and/or postnatal expression of genes involved in lung development along with an impaired alveolarization in female prenatally stressed mice. CONCLUSION: Here we have shown that the fetal origin of asthma is orchestrated by a disrupted airway epithelium and further perpetuated by a predisposed immune system.


Subject(s)
Asthma/immunology , Lung/immunology , Prenatal Exposure Delayed Effects/immunology , Respiratory Mucosa/immunology , Animals , Bone Marrow/immunology , Cells, Cultured , Disease Models, Animal , Female , Immunity/immunology , Inflammation/immunology , Male , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Pregnancy , Respiratory Hypersensitivity/immunology , Tight Junctions/immunology
20.
Anim Sci J ; 91(1): e13357, 2020.
Article in English | MEDLINE | ID: mdl-32219956

ABSTRACT

Tight junctions (TJs) play an important role in intestinal barrier function. TJs in intestinal epithelial cells are composed of different junctional molecules, such as claudin and occludin, and regulate the paracellular permeability of water, ions, and macromolecules in adjacent cells. One of the most important roles of the TJ structure is to provide a physical barrier to luminal inflammatory molecules. Impaired integrity and structure of the TJ barrier result in a forcible activation of immune cells and chronic inflammation in different tissues. According to recent studies, the intestinal TJ barrier could be regulated, as a potential target, by dietary factors to prevent and reduce different inflammatory disorders, although the precise mechanisms underlying the dietary regulation remain unclear. This review summarizes currently available information on the regulation of the intestinal TJ barrier by food components.


Subject(s)
Animal Nutritional Physiological Phenomena , Food , Intestines/cytology , Intestines/physiology , Nutrients , Tight Junctions/physiology , Animals , Body Water/metabolism , Claudins/metabolism , Epithelial Cells/metabolism , Inflammation , Intestines/immunology , Permeability , Tight Junctions/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...