Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 40.042
1.
Zhonghua Bing Li Xue Za Zhi ; 53(6): 541-545, 2024 Jun 08.
Article Zh | MEDLINE | ID: mdl-38825897

Objective: To investigate the clinicopathological features, diagnosis and differential diagnosis of pseudocarcinomatous hyperplasia of the fallopian tubes. Methods: Sixteen cases of pseudocarcinomatous hyperplasia of the fallopian tubes diagnosed at Obstetrics and Gynecology Hospital of Fudan University from January 2011 to January 2024 were collected.The pathological sections were reviewed, the clinical and pathological data were consulted, and immunohistochemical examination was conducted along with follow-up. Results: The patients were aged from 19 to 57 years, with an average age of 41 and a median age of 38. Among the 16 cases, 4 were located in the right fallopian tubes, 6 in the left fallopian tubes, while the remaining cases presented bilaterally. The general manifestations were tubal edema, crispness and purulent secretion in the lumen. Morphologically, the fallopian tube mucosa exhibited a significant infiltration of neutrophils, lymphocytes and plasma cells. The epithelial cells of the fallopian tube displayed evident proliferation, stratification and disorganized arrangement leading to formation of small glandular cavity with back-to-back, fissure-like and sieve-like structures. Immunohistochemical analysis revealed positivity for CK7 and WT1, along with wild-type p53 expression, Ki-67 index ranged from 5% to 20%. During the follow-up period ranging from 1 to 156 months, all the patients remained free of disease. Conclusions: Pseudocarcinomatous hyperplasia of the fallopian tube is a rare non-neoplastic lesion, which can lead to epithelial hyperplasia and atypical hyperplasia. The most important significance of recognizing this lesion lies in avoiding misdiagnosis of fallopian tube cancer during intraoperative and postoperative pathological examination. This ensures that clinicians can administer correct clinical interventions.


Fallopian Tubes , Hyperplasia , Humans , Female , Adult , Hyperplasia/pathology , Middle Aged , Fallopian Tubes/pathology , Fallopian Tubes/metabolism , Diagnosis, Differential , Tumor Suppressor Protein p53/metabolism , Keratin-7/metabolism , Fallopian Tube Neoplasms/pathology , Fallopian Tube Neoplasms/metabolism , Fallopian Tube Neoplasms/surgery , Fallopian Tube Neoplasms/diagnosis , Ki-67 Antigen/metabolism , WT1 Proteins/metabolism , Young Adult , Epithelial Cells/pathology , Epithelial Cells/metabolism , Immunohistochemistry , Fallopian Tube Diseases/pathology , Fallopian Tube Diseases/metabolism , Fallopian Tube Diseases/diagnosis
2.
J Gene Med ; 26(6): e3708, 2024 Jun.
Article En | MEDLINE | ID: mdl-38837511

BACKGROUND: Lysophosphatidic acid (LPA) is a small bioactive lipid which acts as a potent regulator in various tumor progressions through six G-protein-coupled receptors (LPA1-LPA6). Our previous study demonstrated that the LPA-producing enzyme, autotaxin (ATX), was upregulated in esophageal squamous cell carcinoma (ESCC) and ATX high expression levels indicated a poor prognosis. Esophageal squamous cell carcinoma is a type of malignant tumor which originates from epithelial cells. Its progression can be affected by the interaction between cancer cells and normal cells. However, the impact of LPA on the interaction between esophageal epithelial cells and cancer cells in the development of ESCC remains uncertain. METHODS: MTS and Edu assays were performed to determine ESCC cell proliferation in culture medium (CM) derived from LPA-stimulated esophageal epithelial cells (Het-1a). A wound healing assay, transwell migration and an invasion assay were performed to assess the metastatic ability of ESCC cells. Cytokine array analysis was conducted to detect the differentially secreted cytokines in CM. The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were utilized to uncover the pathways and cytokines that are influenced by LPA in ESCC. Immunohistochemical staining was employed to measure the expression of ATX and CCL2 in early-stage ESCC. Quantitative real-time PCR, western blot, enzyme-linked immunosorbent assay and an antibody neutralization assay were employed to measure the mechanism of LPA-mediated communication between epithelial cells and cancer cells. RESULTS: Functional experiments showed that exposing ESCC cancer cells to CM from LPA-treated Het-1a results in promoting proliferation, migration, invasion and epithelial-mesenchymal transition processes. Using cytokine array analysis, we discovered that LPA triggers the release of multiple cytokines from epithelial cells. After screening of the TCGA and GEO databases, CCL2 was identified and found to be correlated with ATX expression in ESCC. Furthermore, CCL2 levels in both mRNA expression and secretion were observed to be upregulated in epithelial cells upon stimulation with LPA. Blocking CCL2 effectively reduced the pro-migration influence of CM derived from LPA-treated Het-1a. Mechanism studies have demonstrated that LPA activated the NF-κB signaling pathway through LPA1/3, ultimately causing an increase in CCL2 expression and secretion in Het-1a. CONCLUSIONS: Our findings, taken together, demonstrate that CM from LPA-treated esophageal epithelial cells plays a significant role in promoting the progression of ESCC, with CCL2 acting as the primary regulator.


Cell Movement , Cell Proliferation , Chemokine CCL2 , Epithelial Cells , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Gene Expression Regulation, Neoplastic , Lysophospholipids , Humans , Lysophospholipids/metabolism , Lysophospholipids/pharmacology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/genetics , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Cell Proliferation/drug effects , Cell Line, Tumor , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Disease Progression , Signal Transduction/drug effects , Esophagus/metabolism , Esophagus/pathology , Esophagus/drug effects , Epithelial-Mesenchymal Transition/drug effects
3.
Biol Pharm Bull ; 47(6): 1119-1122, 2024.
Article En | MEDLINE | ID: mdl-38839363

DNA methylation is a crucial epigenetic modification that regulates gene expression and determines cell fate; however, the triggers that alter DNA methylation levels remain unclear. Recently, we showed that S-nitrosylation of DNA methyltransferase (DNMT) induces DNA hypomethylation and alters gene expression. Furthermore, we identified DBIC, a specific inhibitor of S-nitrosylation of DNMT3B, to suppress nitric oxide (NO)-induced gene alterations. However, it remains unclear how NO-induced DNA hypomethylation regulates gene expression and whether this mechanism is maintained in normal cells and triggers disease-related changes. To address these issues, we focused on carbonic anhydrase 9 (CA9), which is upregulated under nitrosative stress in cancer cells. We pharmacologically evaluated its regulatory mechanisms using human small airway epithelial cells (SAECs) and DBIC. We demonstrated that nitrosative stress promotes the recruitment of hypoxia-inducible factor 1 alpha to the CA9 promoter region and epigenetically induces CA9 expression in SAECs. Our results suggest that nitrosative stress is a key epigenetic regulator that may cause diseases by altering normal cell function.


Carbonic Anhydrase IX , DNA Methylation , Epigenesis, Genetic , Epithelial Cells , Nitric Oxide , Humans , Nitric Oxide/metabolism , Epithelial Cells/metabolism , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrase IX/genetics , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Promoter Regions, Genetic , Cells, Cultured
4.
Wei Sheng Yan Jiu ; 53(3): 441-454, 2024 May.
Article Zh | MEDLINE | ID: mdl-38839586

OBJECTIVE: To investigate the effects of long-term(7 days and 14 days) bisphenol S(BPS) exposure on the ERß-MAPK signaling pathway, hormone secretion phenotype and cell cycle in human normal ovarian epithelial cells IOSE 80 at actual human exposure level. METHODS: Physiologically based pharmacokinetic model combined with BPS levels in the serum of women along the Yangtze River in China was used to determine the dosing concentrations of BPS, and vehicle control and 17 ß-estradiol(E_2) control were used. Complete medium with corresponding concentrations(0, 6.79×10~(-6), 6.79×10~(-4), 6.79×10~(-2), 6.79 µmol/L BPS and 10 nmol/L E_2) was replaced every 2 days. mRNA expressions of estrogen receptor(ERß and GPR30), key genes in MAPK signaling pathway(P38/JNK/ERK signaling pathway) and gonadotropin-releasing hormone-related genes(GnRH-I, GnRH-II and GnRH-R) were measured by qPCR. The ERß-MAPK signaling pathway inhibitors were employed to detect the effect of long-term exposure to BPS on the cell cycle by flow cytometry. Dose-response relationship analysis was performed to calculate the benchmark does lower confidence limits. RESULTS: Compared to the vehicle control, after 7 days exposure to BPS, the ratio of G_2/M phase was significantly increased(P<0.05), and the mRNA expressions of GnRH-I, GnRH-II and GnRH-R were significantly decreased(P<0.05); after 14 days exposure to BPS, the mRNA expressions of ESR2, MAPK3, and MAPK9 were significantly increased(P<0.05), and the mRNA expressions of GnRH-II and GnRH-R were significantly decreased(P<0.05). The GnRH-II mRNA expression level of BPS treatment for 7 days; the G_0/G_1 phase ratio, MAPK3 and MAPK8 mRNA expression level of BPS exposure for 14 days; and the GnRH-I mRNA expression level after BPS treatment for 7 days and 14 days showed a good dose-response relationship but with poor fit. CONCLUSION: Long-term low-dose exposure to BPS may cause cell cycle arrest by activating the ERß-MAPK signaling pathway, and may lead to changes in the hormone secretion of IOSE 80 cells.


Epithelial Cells , Estrogen Receptor beta , MAP Kinase Signaling System , Ovary , Phenols , Sulfones , Humans , Phenols/toxicity , Female , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Estrogen Receptor beta/metabolism , Estrogen Receptor beta/genetics , MAP Kinase Signaling System/drug effects , Ovary/drug effects , Ovary/metabolism , Sulfones/toxicity , Cell Line
5.
Iran J Allergy Asthma Immunol ; 23(2): 211-219, 2024 Apr 07.
Article En | MEDLINE | ID: mdl-38822515

Asthma is a chronic respiratory disease that is characterized by airway inflammation, excessive mucus production, and airway remodeling. Prevention and treatment for asthma is an urgent issue in clinical studies. In recent years, N6-methyladenosine methylation (m6A) has emerged as a promising regulatory approach involved in multiple diseases. ALKBH5 (alkB homolog 5) is a demethylase widely studied in disease pathologies. This work aimed to explore the regulatory mechanisms underlying the ALKBH5-regulated asthma. We established an interleukin-13 (IL-13)-stimulated cell model to mimic the in vitro inflammatory environment of asthma. ALKBH5 knockdown in bronchial epithelial cells was performed using siRNAs, and the knockdown efficacy was analyzed by quantitative PCR (qPCR). Cell viability and proliferation were measured by cell counting kit 8 (CCK-8) and colony formation assay. The ferroptosis was assessed by measuring the total iron, Fe2+, lipid reactive oxygen species (ROS), malondialdehyde (MDA), and superoxide dismutase (SOD) levels. The enrichment of N6-methyladenosine methylation (m6A) modification was detected by the MeRIP assay. Knockdown of ALKBH5 significantly elevated the survival and colony formation ability of bronchial epithelial cells in the IL-13 induction model. The levels of total iron, Fe2+, lipid ROS, and MDA were remarkedly elevated, and the SOD level was reduced in IL-13-induced bronchial epithelial cells, and depletion of ALKBH5 reversed these effects. Knockdown of ALKBH5 elevated the enrichment of m6A modification and expression of glutathione peroxidase 4 (GPX4). Knockdown of GPX4 abolished the pro-proliferation and anti-ferroptosis effects of siALKBH5. Knockdown of ALKBH5 improved the proliferation of bronchial epithelial cells and alleviated cell ferroptosis.


Adenosine , AlkB Homolog 5, RNA Demethylase , Asthma , AlkB Homolog 5, RNA Demethylase/metabolism , AlkB Homolog 5, RNA Demethylase/genetics , Asthma/genetics , Asthma/metabolism , Asthma/pathology , Humans , Adenosine/analogs & derivatives , Adenosine/metabolism , Cell Proliferation/genetics , Methylation , Disease Progression , Cell Line , Ferroptosis/genetics , Epithelial Cells/metabolism , Down-Regulation , Bronchi/pathology , Bronchi/metabolism , Gene Knockdown Techniques , Cell Survival/genetics
6.
Elife ; 122024 Jun 05.
Article En | MEDLINE | ID: mdl-38836551

Tuft cells are a group of rare epithelial cells that can detect pathogenic microbes and parasites. Many of these cells express signaling proteins initially found in taste buds. It is, however, not well understood how these taste signaling proteins contribute to the response to the invading pathogens or to the recovery of injured tissues. In this study, we conditionally nullified the signaling G protein subunit Gγ13 and found that the number of ectopic tuft cells in the injured lung was reduced following the infection of the influenza virus H1N1. Furthermore, the infected mutant mice exhibited significantly larger areas of lung injury, increased macrophage infiltration, severer pulmonary epithelial leakage, augmented pyroptosis and cell death, greater bodyweight loss, slower recovery, worsened fibrosis and increased fatality. Our data demonstrate that the Gγ13-mediated signal transduction pathway is critical to tuft cells-mediated inflammation resolution and functional repair of the damaged lungs.To our best knowledge, it is the first report indicating subtype-specific contributions of tuft cells to the resolution and recovery.


Influenza A Virus, H1N1 Subtype , Signal Transduction , Animals , Mice , Influenza A Virus, H1N1 Subtype/physiology , Orthomyxoviridae Infections , Lung Injury/metabolism , Lung/pathology , Inflammation , Epithelial Cells/metabolism , Mice, Knockout , Disease Models, Animal
7.
Nat Commun ; 15(1): 4775, 2024 Jun 05.
Article En | MEDLINE | ID: mdl-38839750

The metal ion transporter SLC39A8 is associated with physiological traits and diseases, including blood manganese (Mn) levels and inflammatory bowel diseases (IBD). The mechanisms by which SLC39A8 controls Mn homeostasis and epithelial integrity remain elusive. Here, we generate Slc39a8 intestinal epithelial cell-specific-knockout (Slc39a8-IEC KO) mice, which display markedly decreased Mn levels in blood and most organs. Radiotracer studies reveal impaired intestinal absorption of dietary Mn in Slc39a8-IEC KO mice. SLC39A8 is localized to the apical membrane and mediates 54Mn uptake in intestinal organoid monolayer cultures. Unbiased transcriptomic analysis identifies alkaline ceramidase 1 (ACER1), a key enzyme in sphingolipid metabolism, as a potential therapeutic target for SLC39A8-associated IBDs. Importantly, treatment with an ACER1 inhibitor attenuates colitis in Slc39a8-IEC KO mice by remedying barrier dysfunction. Our results highlight the essential roles of SLC39A8 in intestinal Mn absorption and epithelial integrity and offer a therapeutic target for IBD associated with impaired Mn homeostasis.


Alkaline Ceramidase , Cation Transport Proteins , Inflammatory Bowel Diseases , Intestinal Mucosa , Manganese , Mice, Knockout , Animals , Cation Transport Proteins/metabolism , Cation Transport Proteins/genetics , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Manganese/metabolism , Mice , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Alkaline Ceramidase/metabolism , Alkaline Ceramidase/genetics , Humans , Mice, Inbred C57BL , Homeostasis , Male , Colitis/metabolism , Colitis/genetics , Colitis/pathology , Intestinal Absorption , Epithelial Cells/metabolism
8.
Sci Rep ; 14(1): 12879, 2024 06 05.
Article En | MEDLINE | ID: mdl-38839896

Paneth cells (PCs), a subset of intestinal epithelial cells (IECs) found at the base of small intestinal crypts, play an essential role in maintaining intestinal homeostasis. Altered PCs function is associated with diverse intestinal pathologies, including ileal Crohn's disease (CD). CD patients with ileal involvement have been previously demonstrated to display impairment in PCs and decreased levels of anti-microbial peptides. Although the immunosuppressive drug Azathioprine (AZA) is widely used in CD therapy, the impact of AZA on IEC differentiation remains largely elusive. In the present study, we hypothesized that the orally administered drug AZA also exerts its effect through modulation of the intestinal epithelium and specifically via modulation of PC function. AZA-treated CD patients exhibited an ileal upregulation of AMPs on both mRNA and protein levels compared to non-AZA treated patients. Upon in vitro AZA stimulation, intestinal epithelial cell line MODE-K exhibited heightened expression levels of PC marker in concert with diminished cell proliferation but boosted mitochondrial OXPHOS activity. Moreover, differentiation of IECs, including PCs differentiation, was boosted in AZA-treated murine small intestinal organoids and was associated with decreased D-glucose consumption and decreased growth rates. Of note, AZA treatment strongly decreased Lgr5 mRNA expression as well as Ki67 positive cells. Further, AZA restored dysregulated PCs associated with mitochondrial dysfunction. AZA-dependent inhibition of IEC proliferation is accompanied by boosted mitochondria function and IEC differentiation into PC.


Azathioprine , Cell Differentiation , Crohn Disease , Intestinal Mucosa , Paneth Cells , Crohn Disease/drug therapy , Crohn Disease/pathology , Crohn Disease/metabolism , Azathioprine/pharmacology , Paneth Cells/metabolism , Paneth Cells/drug effects , Paneth Cells/pathology , Humans , Cell Differentiation/drug effects , Animals , Mice , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Female , Male , Ileum/drug effects , Ileum/metabolism , Ileum/pathology , Adult , Organoids/drug effects , Organoids/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Cell Proliferation/drug effects , Middle Aged , Cell Line , Severity of Illness Index
9.
Sci Rep ; 14(1): 12744, 2024 06 03.
Article En | MEDLINE | ID: mdl-38830931

Transforming growth factor ß (TGF-ß) is implicated in both mesothelial-to-mesenchymal transition (MMT) and cellular senescence of human peritoneal mesothelial cells (HPMCs). We previously showed that senescent HPMCs could spontaneously acquire some phenotypic features of MMT, which in young HPMCs were induced by TGF-ß. Here, we used electron microscopy, as well as global gene and protein profiling to assess in detail how exposure to TGF-ß impacts on young and senescent HPMCs in vitro. We found that TGF-ß induced structural changes consistent with MMT in young, but not in senescent HPMCs. Of all genes and proteins identified reliably in HPMCs across all treatments and states, 4,656 targets represented overlapping genes and proteins. Following exposure to TGF-ß, 137 proteins and 46 transcripts were significantly changed in young cells, compared to 225 proteins and only 2 transcripts in senescent cells. Identified differences between young and senescent HPMCs were related predominantly to wound healing, integrin-mediated signalling, production of proteases and extracellular matrix components, and cytoskeleton structure. Thus, the response of senescent HPMCs to TGF-ß differs or is less pronounced compared to young cells. As a result, the character and magnitude of the postulated contribution of HPMCs to TGF-ß-induced peritoneal remodelling may change with cell senescence.


Cellular Senescence , Epithelial Cells , Peritoneum , Transforming Growth Factor beta , Humans , Cellular Senescence/drug effects , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Peritoneum/cytology , Peritoneum/metabolism , Epithelial-Mesenchymal Transition/drug effects , Cells, Cultured , Epithelium/metabolism , Epithelium/drug effects , Signal Transduction/drug effects , Gene Expression Profiling
10.
Ren Fail ; 46(2): 2359638, 2024 Dec.
Article En | MEDLINE | ID: mdl-38832484

Emerging data have revealed that damage to tubular epithelial cell is a driving force in the progression of diabetic kidney disease (DKD). However, the specific mechanisms by which lipotoxicity contributes to the injury of these cells, thereby influencing the development of DKD, are yet to be fully understood. Here, we analyzed the GSE 30529 microarray datasets of human tubulointerstitial tissue samples from the Gene Expression Omnibus database (GEO). Concurrently, we conducted RNA-sequencing on palmitic acid (PA)-treated human renal proximal tubule epithelial cells (HK2 cells). After normalization, the differentially expressed genes (DEGs) were screened by R software and gene ontology (GO) enrichment analysis was conducted, and lysosomal-associated protein transmembrane 5 (LAPTM5) was finally selected. Our findings indicate that the expression of LAPTM5 was obviously increased in DKD patients, and the correlation between LAPTM5, and other clinical parameters of DKD was analyzed using the Spearman correlation analysis. The potential of LAPTM5 as a prognostic biomarker for DKD was further consolidated through receiver operating characteristic (ROC) analysis. To further verify the function of LAPTM5, we established mouse or in vitro systems mimicking DKD. The results showed that a consistent upregulation of LAPTM5, which was also found to be linked with inflammatory mediators within the context of DKD. Additionally, LAPTM5 silencing significantly downregulated mRNA expression of inflammatory factors in PA-treated HK2 cells. These results indicate that LAPTM5 is a potential biomarker and therapeutic treatment target for DKD. This discovery paves the way for future research and development of targeted interventions aimed at mitigating the progression of this prevalent condition.


Computational Biology , Diabetic Nephropathies , Membrane Proteins , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/genetics , Diabetic Nephropathies/etiology , Diabetic Nephropathies/pathology , Humans , Animals , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Cell Line , Palmitic Acid/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Male , Mice, Inbred C57BL , Up-Regulation , Biomarkers/metabolism
11.
J Nanobiotechnology ; 22(1): 236, 2024 May 10.
Article En | MEDLINE | ID: mdl-38724995

Increased proinflammatory cytokines and infiltration of inflammatory cells in the stroma are important pathological features of type IIIA chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS-A), and the interaction between stromal cells and other cells in the inflammatory microenvironment is closely related to the inflammatory process of CP/CPPS-A. However, the interaction between stromal and epithelial cells remains unclear. In this study, inflammatory prostate epithelial cells (PECs) released miR-203a-3p-rich exosomes and facilitated prostate stromal cells (PSCs) inflammation by upregulating MCP-1 expression. Mechanistically, DUSP5 was identified as a novel target gene of miR-203a-3p and regulated PSCs inflammation through the ERK1/2/MCP-1 signaling pathway. Meanwhile, the effect of exosomes derived from prostatic fluids of CP/CPPS-A patients was consistent with that of exosomes derived from inflammatory PECs. Importantly, we demonstrated that miR-203a-3p antagomirs-loaded exosomes derived from PECs targeted the prostate and alleviated prostatitis by inhibiting the DUSP5-ERK1/2 pathway. Collectively, our findings provide new insights into underlying the interaction between PECs and PSCs in CP/CPPS-A, providing a promising therapeutic strategy for CP/CPPS-A.


Epithelial Cells , Exosomes , MicroRNAs , Prostatitis , Stromal Cells , Male , Exosomes/metabolism , Prostatitis/genetics , Prostatitis/pathology , Prostatitis/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Animals , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/metabolism , Prostate/pathology , Prostate/metabolism , Pelvic Pain , Inflammation/genetics , Inflammation/pathology , Mice , MAP Kinase Signaling System
12.
Cells ; 13(9)2024 May 02.
Article En | MEDLINE | ID: mdl-38727316

Epithelial-mesenchymal transition (EMT) is a process during which epithelial cells lose epithelial characteristics and gain mesenchymal features. Here, we used several cell models to study migratory activity and redistribution of cell-cell adhesion proteins in cells in different EMT states: EGF-induced EMT of epithelial IAR-20 cells; IAR-6-1 cells with a hybrid epithelial-mesenchymal phenotype; and their more mesenchymal derivatives, IAR-6-1-DNE cells lacking adherens junctions. In migrating cells, the cell-cell adhesion protein α-catenin accumulated at the leading edges along with ArpC2/p34 and α-actinin. Suppression of α-catenin shifted cell morphology from fibroblast-like to discoid and attenuated cell migration. Expression of exogenous α-catenin in MDA-MB-468 cells devoid of α-catenin drastically increased their migratory capabilities. The Y654 phosphorylated form of ß-catenin was detected at integrin adhesion complexes (IACs). Co-immunoprecipitation studies indicated that α-catenin and pY654-ß-catenin were associated with IAC proteins: vinculin, zyxin, and α-actinin. Taken together, these data suggest that in cells undergoing EMT, catenins not participating in assembly of adherens junctions may affect cell migration.


Actin Cytoskeleton , Cell Movement , Epithelial-Mesenchymal Transition , alpha Catenin , Humans , Actin Cytoskeleton/metabolism , alpha Catenin/metabolism , beta Catenin/metabolism , Vinculin/metabolism , Adherens Junctions/metabolism , Cell Adhesion , Actinin/metabolism , Cell Line, Tumor , Zyxin/metabolism , Phosphorylation , Integrins/metabolism , Animals , Epithelial Cells/metabolism
13.
Mol Biol Rep ; 51(1): 637, 2024 May 10.
Article En | MEDLINE | ID: mdl-38727927

BACKGROUND: Retinal pigment epithelial cells (RPECs) are a type of retinal cells that structurally and physiologically support photoreceptors. However, hyperglycemia has been shown to play a critical role in the progression of diabetic retinopathy (DR), which is one of the leading causes of vision impairment. In the diabetic eye, the high glucose environment damages RPECs via the induction of oxidative stress, leading to the release of excess reactive oxygen species (ROS) and triggering apoptosis. In this study, we aim to investigate the antioxidant mechanism of Vitamin C in reducing hyperglycemia-induced stress and whether this mechanism can preserve the function of RPECs. METHODS AND RESULTS: ARPE-19 cells were treated with high glucose in the presence or absence of Vitamin C. Cell viability was measured by MTT assay. Cleaved poly ADP-ribose polymerase (PARP) was used to identify apoptosis in the cells. ROS were detected by the DCFH-DA reaction. The accumulation of sorbitol in the aldose reductase (AR) polyol pathway was determined using the sorbitol detection assay. Primary mouse RPECs were isolated from adult mice and identified by Rpe65 expression. The mitochondrial damage was measured by mitochondrial membrane depolarization. Our results showed that high glucose conditions reduce cell viability in RPECs while Vitamin C can restore cell viability, compared to the vehicle treatment. We also demonstrated that Vitamin C reduces hyperglycemia-induced ROS production and prevents cell apoptosis in RPECs in an AR-independent pathway. CONCLUSIONS: These results suggest that Vitamin C is not only a nutritional necessity but also an adjuvant that can be combined with AR inhibitors for alleviating hyperglycemic stress in RPECs.


Apoptosis , Ascorbic Acid , Cell Survival , Glucose , Hyperglycemia , Oxidative Stress , Reactive Oxygen Species , Retinal Pigment Epithelium , Ascorbic Acid/pharmacology , Ascorbic Acid/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/drug effects , Hyperglycemia/metabolism , Hyperglycemia/drug therapy , Hyperglycemia/complications , Animals , Reactive Oxygen Species/metabolism , Mice , Oxidative Stress/drug effects , Apoptosis/drug effects , Cell Survival/drug effects , Glucose/metabolism , Humans , Cell Line , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/drug therapy , Antioxidants/pharmacology , Antioxidants/metabolism , Mitochondria/metabolism , Mitochondria/drug effects
14.
Transl Vis Sci Technol ; 13(5): 3, 2024 May 01.
Article En | MEDLINE | ID: mdl-38696180

Purpose: The biosynthetic Symatix membrane (SM) was developed to replace fresh human amniotic membrane (hAM) in ocular surgical applications. The purpose of this study was to test the biocompatibility of the SM with human limbus-derived epithelial cells with regard to their physical and biological properties. Methods: Different physical properties of SM were tested ex vivo by simulation on human corneas. In vitro, primary limbal epithelial cells from limbal explants were used to test biological properties such as cell migration, proliferation, metabolic activity, and limbal epithelial cell markers on the SM, hAM, and freeze-dried amniotic membrane (FDAM). Results: The surgical handleability of the SM was equivalent to that of the hAM. Ultrastructural and histological studies demonstrated that epithelial cells on the SM had the typical tightly apposed, polygonal, corneal epithelial cell morphology. The epithelial cells were well stratified on the SM, unlike on the hAM and FDAM. Rapid wound healing occurred on the SM within 3 days. Immunofluorescence studies showed positive expression of CK-19, Col-1, laminin, ZO-1, FN, and p-63 on the SM, plastic, and FDAM compared to positive expression of ZO-1, Col-1, laminin, FN, and p63 and negative expression of CK-19 in the hAM. Conclusions: These results indicate that the SM is a better substrate for limbal epithelial cell migration, proliferation, and tight junction formation. Altogether, the SM can provide a suitable alternative to the hAM for surgical application in sight-restoring operations. Translational Relevance: The hAM, currently widely used in ocular surface surgery, has numerous variations and limitations. The biocompatibility of corneal epithelial cells with the SM demonstrated in this study suggests that it can be a viable substitute for the hAM.


Amnion , Cell Movement , Cell Proliferation , Humans , Amnion/metabolism , Cells, Cultured , Limbus Corneae/metabolism , Limbus Corneae/cytology , Epithelium, Corneal/metabolism , Epithelium, Corneal/cytology , Wound Healing/physiology , Epithelial Cells/metabolism , Ophthalmologic Surgical Procedures/methods , Laminin/metabolism , Zonula Occludens-1 Protein/metabolism
15.
Front Immunol ; 15: 1342350, 2024.
Article En | MEDLINE | ID: mdl-38720901

Dyslipidemia is the most prevalent independent risk factor for patients with chronic kidney disease (CKD). Lipid-induced NLRP3 inflammasome activation in kidney-resident cells exacerbates renal injury by causing sterile inflammation. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that modulates the cellular redox balance; however, the exact role of Nrf2 signaling and its regulation of the NLRP3 inflammasome in hyperlipidemia-induced kidney injury are poorly understood. In this study, we demonstrated that activation of the mtROS-NLRP3 inflammasome pathway is a critical contributor to renal tubular epithelial cell (RTEC) apoptosis under hyperlipidemia. In addition, the Nrf2/ARE signaling pathway is activated in renal tubular epithelial cells under hyperlipidemia conditions both in vivo and in vitro, and Nrf2 silencing accelerated palmitic acid (PA)-induced mtROS production, mitochondrial injury, and NLRP3 inflammasome activation. However, the activation of Nrf2 with tBHQ ameliorated mtROS production, mitochondrial injury, NLRP3 inflammasome activation, and cell apoptosis in PA-induced HK-2 cells and in the kidneys of HFD-induced obese rats. Furthermore, mechanistic studies showed that the potential mechanism of Nrf2-induced NLRP3 inflammasome inhibition involved reducing mtROS generation. Taken together, our results demonstrate that the Nrf2/ARE signaling pathway attenuates hyperlipidemia-induced renal injury through its antioxidative and anti-inflammatory effects through the downregulation of mtROS-mediated NLRP3 inflammasome activation.


Epithelial Cells , Hyperlipidemias , Inflammasomes , Kidney Tubules , NF-E2-Related Factor 2 , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction , NF-E2-Related Factor 2/metabolism , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Hyperlipidemias/metabolism , Hyperlipidemias/complications , Hyperlipidemias/immunology , Epithelial Cells/metabolism , Rats , Humans , Kidney Tubules/pathology , Kidney Tubules/metabolism , Male , Cell Line , Apoptosis , Antioxidant Response Elements , Mitochondria/metabolism , Disease Models, Animal , Rats, Sprague-Dawley
16.
Sci Adv ; 10(19): eadi8433, 2024 May 10.
Article En | MEDLINE | ID: mdl-38718115

Cell deformability is an essential determinant for tissue-scale mechanical nature, such as fluidity and rigidity, and is thus crucial for tissue homeostasis and stable developmental processes. However, large-scale simulations of deformable cells have been restricted to those of polygonal-shaped cells, limiting our understanding of populations of arbitrarily deformable cells, such as mesenchymal, amoeboid cells, and nonconfluent epithelial cells. Here, we present an efficient approach for simulating large populations of nonpolygonally deformable cells with considerably higher computational efficiency than existing methods. Using the method, we demonstrate that the densely packed active cell population interacting via excluded volume interactions exhibits a fluid-to-fluid transition. An experimentally measurable index of topological defects, defined using the number of neighboring cells, is also proposed to characterize this transition. This study provides a flexible approach to tissue-scale cell population and a broader perspective on the biological fluid phases.


Models, Biological , Phase Transition , Humans , Cell Shape , Computer Simulation , Epithelial Cells/metabolism , Epithelial Cells/cytology
17.
Nat Commun ; 15(1): 3666, 2024 Apr 30.
Article En | MEDLINE | ID: mdl-38693120

Respiratory viral infection increases host susceptibility to secondary bacterial infections, yet the precise dynamics within airway epithelia remain elusive. Here, we elucidate the pivotal role of CD47 in the airway epithelium during bacterial super-infection. We demonstrated that upon influenza virus infection, CD47 expression was upregulated and localized on the apical surface of ciliated cells within primary human nasal or bronchial epithelial cells. This induced CD47 exposure provided attachment sites for Staphylococcus aureus, thereby compromising the epithelial barrier integrity. Through bacterial adhesion assays and in vitro pull-down assays, we identified fibronectin-binding proteins (FnBP) of S. aureus as a key component that binds to CD47. Furthermore, we found that ciliated cell-specific CD47 deficiency or neutralizing antibody-mediated CD47 inactivation enhanced in vivo survival rates. These findings suggest that interfering with the interaction between airway epithelial CD47 and pathogenic bacterial FnBP holds promise for alleviating the adverse effects of super-infection.


CD47 Antigen , Epithelial Cells , Staphylococcal Infections , Staphylococcus aureus , Superinfection , CD47 Antigen/metabolism , CD47 Antigen/genetics , Humans , Animals , Superinfection/microbiology , Mice , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/virology , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Influenza, Human/metabolism , Influenza, Human/immunology , Influenza, Human/virology , Bacterial Adhesion , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Respiratory Mucosa/virology , Mice, Inbred C57BL , Bronchi/metabolism , Bronchi/cytology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Mice, Knockout , Influenza A Virus, H1N1 Subtype
18.
BMC Genomics ; 25(1): 520, 2024 May 27.
Article En | MEDLINE | ID: mdl-38802796

BACKGROUND: Increasing evidence points to an active role of oviductal extracellular vesicles (oEVs) in the early embryo-maternal dialogue. However, it remains unclear whether oEVs contribute to the recognition of the presence of embryos and their quality in the oviduct. Hence, we examined whether the molecular cargo of oEVs secreted by bovine oviduct epithelial cells (BOEC) differs depending on the presence of good (≥ 8 cells, G) or poor (< 8 cells, P) quality embryos. In addition, differences in RNA profiles between G and P embryos were analyzed in attempt to distinguish oEVs and embryonic EVs cargos. METHODS: For this purpose, primary BOEC were co-cultured with in vitro produced embryos (IVP) 53 h post fertilization as follows: BOEC with G embryos (BGE); BOEC with P embryos (BPE); G embryos alone (GE); P embryos alone (PE); BOEC alone (B) and medium control (M). After 24 h of co-culture, conditioned media were collected from all groups and EVs were isolated and characterized. MicroRNA profiling of EVs and embryos was performed by small RNA-sequencing. RESULTS: In EVs, 84 miRNAs were identified, with 8 differentially abundant (DA) miRNAs for BGE vs. B and 4 for BPE vs. B (P-value < 0.01). In embryos, 187 miRNAs were identified, with 12 DA miRNAs for BGE vs. BPE, 3 for G vs. P, 8 for BGE vs. GE, and 11 for BPE vs. PE (P-value < 0.01). CONCLUSIONS: These results indicated that oEVs are involved in the oviductal-embryo recognition and pointed to specific miRNAs with signaling and supporting roles during early embryo development.


Embryo, Mammalian , Extracellular Vesicles , MicroRNAs , Oviducts , Animals , Extracellular Vesicles/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Female , Cattle , Embryo, Mammalian/metabolism , Oviducts/metabolism , Oviducts/cytology , Epithelial Cells/metabolism , Coculture Techniques , Fallopian Tubes/metabolism , Fallopian Tubes/cytology
19.
J Vis Exp ; (207)2024 May 10.
Article En | MEDLINE | ID: mdl-38801260

The squamous epithelium of the esophagus is directly exposed to the environment, continuously facing foreign antigens, including food antigens and microbes. Maintaining the integrity of the epithelial barrier is critical for preventing infections and avoiding inflammation caused by harmless food-derived antigens. This article provides simplified protocols for generating human esophageal organoids and air-liquid interface cultures from patient biopsies to study the epithelial compartment of the esophagus in the context of tissue homeostasis and disease. These protocols have been significant scientific milestones in the last decade, describing three-dimensional organ-like structures from patient-derived primary cells, organoids, and air-liquid interface cultures. They offer the possibility to investigate the function of specific cytokines, growth factors, and signaling pathways in the esophageal epithelium within a three-dimensional framework while maintaining the phenotypic and genetic properties of the donor. Organoids provide information on tissue microarchitecture by assessing the transcriptome and proteome after cytokine stimulation. In contrast, air-liquid interface cultures allow the assessment of the epithelial barrier integrity through transepithelial resistance (TEER) or macromolecule flux measurements. Combining these organoids and air-liquid interface cultures is a powerful tool to advance research in impaired esophageal epithelial barrier conditions.


Eosinophilic Esophagitis , Organoids , Eosinophilic Esophagitis/pathology , Eosinophilic Esophagitis/metabolism , Humans , Organoids/pathology , Organoids/metabolism , Cell Culture Techniques, Three Dimensional/methods , Esophagus/pathology , Esophagus/cytology , Cell Culture Techniques/methods , Epithelial Cells/metabolism , Epithelial Cells/pathology
20.
Invest Ophthalmol Vis Sci ; 65(5): 24, 2024 May 01.
Article En | MEDLINE | ID: mdl-38748430

Purpose: Hydrogels derived from decellularized tissues are promising biomaterials in tissue engineering, but their rapid biodegradation can hinder in vitro cultivation. This study aimed to retard biodegradation of a hydrogel derived from porcine decellularized lacrimal glands (dLG-HG) by crosslinking with genipin to increase the mechanical stability without affecting the function and viability of lacrimal gland (LG)-associated cells. Methods: The effect of different genipin concentrations on dLG-HG stiffness was measured rheologically. Cell-dependent biodegradation was quantified over 10 days, and the impact on matrix metalloproteinase (MMP) activity was quantified by gelatin and collagen zymography. The viability of LG epithelial cells (EpCs), mesenchymal stem cells (MSCs), and endothelial cells (ECs) cultured on genipin-crosslinked dLG-HG was assessed after 10 days, and EpC secretory activity was analyzed by ß-hexosaminidase assay. Results: The 0.5-mM genipin increased the stiffness of dLG-HG by about 46%, and concentrations > 0.25 mM caused delayed cell-dependent biodegradation and reduced MMP activity. The viability of EpCs, MSCs, and ECs was not affected by genipin concentrations of up to 0.5 mM after 10 days. Moreover, up to 0.5-mM genipin did not negatively affect EpC secretory activity compared to control groups. Conclusions: A concentration of 0.5-mM genipin increased dLG-HG stiffness, and 0.25-mM genipin was sufficient to prevent MMP-dependent degradation. Importantly, concentrations of up to 0.5-mM genipin did not compromise the viability of LG-associated cells or the secretory activity of EpCs. Thus, crosslinking with genipin improves the properties of dLG-HG for use as a substrate in LG tissue engineering.


Cell Survival , Cross-Linking Reagents , Hydrogels , Iridoids , Tissue Engineering , Animals , Iridoids/pharmacology , Iridoids/metabolism , Swine , Tissue Engineering/methods , Cross-Linking Reagents/pharmacology , Cells, Cultured , Mesenchymal Stem Cells/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Biocompatible Materials
...