Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40.008
Filter
1.
Sci Adv ; 10(19): eadi8433, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38718115

ABSTRACT

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.


Subject(s)
Models, Biological , Phase Transition , Humans , Cell Shape , Computer Simulation , Epithelial Cells/metabolism , Epithelial Cells/cytology
2.
Transl Vis Sci Technol ; 13(5): 3, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38696180

ABSTRACT

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.


Subject(s)
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
3.
Biomed Res Int ; 2024: 8544837, 2024.
Article in English | MEDLINE | ID: mdl-38803515

ABSTRACT

The loss of RAB25 expression-RAS superfamily of GTPase characteristic of numerous breast cancers-corresponds with H-RAS point mutations, particularly in triple-negative breast cancers (TNBC), a subtype associated with a poor prognosis. To address the poorly understood factors dictating the progression of TNBC tumors, we examine the cooperative effects that loss of RAB25 expression in human mammary epithelial cell (HMEC) lines with H-RAS mutations confers in tumorigenesis. HMECs were immortalized by transduction with LXSN CDK4 R24C, a mutant form of cyclin-dependent kinase, followed by transduction with hTERT, a catalytic subunit of the telomerase enzyme. We found that with the loss of RAB25 and overexpression of mutant H-RAS61L, immortal HMECs transformed toward anchorage-independent growth and acquired an increased ability to migrate. Furthermore, cells express low CD24, high CD44, and low claudin levels, indicating stem-like properties upon transformation. Besides, loss of RAB25 and overexpression of H-RAS61L resulted in increased expression of transcription factors Snail and Slug that drive these cells to lose E-cadherin and undergo epithelial-mesenchymal transition (EMT). This study confirms that loss of RAB25 and overexpression of mutant H-RAS can drive HMECs toward a mesenchymal stem-like state. Our findings reveal that RAB25 functions as a tumor suppressor gene, and loss of RAB25 could serve as a novel biomarker of the claudin-low type of TNBC.


Subject(s)
Cell Transformation, Neoplastic , Claudins , Epithelial Cells , Epithelial-Mesenchymal Transition , rab GTP-Binding Proteins , Humans , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Claudins/genetics , Claudins/metabolism , Female , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Oncogenes/genetics , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Mutation/genetics
4.
Cell Death Dis ; 15(5): 365, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806451

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is one of the main causes of peritoneal fibrosis. However, the pathophysiological mechanisms of EMT, specifically its relationship with autophagy, are still unknown. This study aimed to evaluate the role of autophagy in transforming growth factor-beta 1 (TGF-ß1)-induced EMT in human peritoneal mesothelial cells (HPMCs). Primary cultured HPMCs were treated with TGF-ß1 (2 and 5 ng/mL) and changes in autophagy markers and the relationship between autophagy and EMT were evaluated. We also identified changes in EMT- and autophagy-related signaling pathways after autophagy and NADPH oxidase 4 (NOX4) inhibition. TGF-ß1 increased the generation of NOX4 and reactive oxygen species (ROS) in HPMCs, resulting in mitochondrial damage. Treatment with GKT137831 (20 µM), a NOX1/4 inhibitor, reduced ROS in the mitochondria of HPMC cells and reduced TGF-ß1-induced mitochondrial damage. Additionally, the indirect inhibition of autophagy by GKT137831 (20 µM) downregulated TGF-ß1-induced EMT, whereas direct inhibition of autophagy using 3-methyladenine (3-MA) (2 mM) or autophagy-related gene 5 (ATG5) gene silencing decreased the TGF-ß1-induced EMT in HPMCs. The suppressor of mothers against decapentaplegic 2/3 (Smad2/3), autophagy-related phosphoinositide 3-kinase (PI3K) class III, and protein kinase B (Akt) pathways, and mitogen-activated protein kinase (MAPK) signaling pathways, such as extracellular signal-regulated kinase (ERK) and P38, were involved in TGF-ß1-induced EMT. Autophagy and NOX4 inhibition suppressed the activation of these signaling pathways. Direct inhibition of autophagy and its indirect inhibition through the reduction of mitochondrial damage by upstream NOX4 inhibition reduced EMT in HPMCs. These results suggest that autophagy could serve as a therapeutic target for the prevention of peritoneal fibrosis in patients undergoing peritoneal dialysis.


Subject(s)
Autophagy , Epithelial Cells , Epithelial-Mesenchymal Transition , NADPH Oxidase 4 , Oxidative Stress , Reactive Oxygen Species , Signal Transduction , Transforming Growth Factor beta1 , Humans , Epithelial-Mesenchymal Transition/drug effects , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta1/metabolism , Autophagy/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Signal Transduction/drug effects , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Epithelial Cells/pathology , Mitochondria/metabolism , Mitochondria/drug effects , Peritoneum/pathology , Pyrazolones , Pyridones
5.
Reprod Domest Anim ; 59(5): e14567, 2024 May.
Article in English | MEDLINE | ID: mdl-38798178

ABSTRACT

In this study, Holstein dairy cows raised in Ningxia were selected as the research object. Mammary epithelial cells (BMECs) were extracted from the milk of eight Holstein cows with significantly different milk fat expression rates and transcribed for sequencing. Bioinformatics analysis was used to analyse the correlation of fat milk percentage, and the critical miR-2285f regulating milk fat was screened out. The target gene binding sites were predicted, and 293T cells and mammary epithelial cells were used as miRNA and target gene models for functional verification in vitro. The tissue difference of miR-2285f Holstein cows was quantitatively analysed by transfecting miR-2285f mimic and inhibitor. Assay (dual luciferase reporter gene assay) and quantitative real-time PCR (quantitative real-time PCR, qRT-PCR), triglyceride (TAG) detection, oil red O detection of lipid droplets, Western Blot assay, Edu and Flow cytometry, The molecular regulatory effects of miR-2285f and target gene MAP2K2 on milk fat metabolism of Holstein dairy cows were studied. The wild-type vector and mutant vector of map2k2-3'utr were constructed, and double luciferase reporting experiments were conducted to verify that MAP2K2 was one of the target genes of miR-2285f. According to qRT-PCR and Western Blot analysis, miR-2285f mainly regulates the expression of MAP2K2 protein in BMECs at the translation level. Bta-miR-2285f can promote cell proliferation and slow cell apoptosis by regulating MAP2K2. Bta-miR-2285f can promote triglyceride (TAG) and lipid droplet accumulation in mammary epithelial cells by targeting MAP2K2. Bta-miR-2285f can regulate protein levels of fat milk marker gene PPARG by targeting MAP2K2. In conclusion, miR-2285f can target the expression of the MAP2K2 gene, promote the proliferation of dairy mammary epithelial cells, inhibit cell apoptosis and regulate the milk fat metabolism in dairy mammary epithelial cells. The results of this study revealed the function of miR-2285f in regulating the differential expression of fat milk in Holstein dairy cows at the cellular level. They provided a theoretical and experimental basis for analysing the regulation network of milk fat synthesis of Holstein dairy cows and the molecular breeding of dairy cows.


Subject(s)
Epithelial Cells , Mammary Glands, Animal , MicroRNAs , Milk , Animals , Cattle , MicroRNAs/metabolism , MicroRNAs/genetics , Female , Milk/chemistry , Mammary Glands, Animal/metabolism , Epithelial Cells/metabolism , MAP Kinase Kinase Kinase 2/metabolism , MAP Kinase Kinase Kinase 2/genetics , Lipid Metabolism , Triglycerides/metabolism , Apoptosis , Humans , Gene Expression Regulation , Cell Proliferation
6.
J Vis Exp ; (207)2024 May 10.
Article in English | MEDLINE | ID: mdl-38801260

ABSTRACT

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.


Subject(s)
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
7.
Biomolecules ; 14(5)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38785926

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is a deadly consequence of radiation exposure to the esophagus. ESCC arises from esophageal epithelial cells that undergo malignant transformation and features a perturbed squamous cell differentiation program. Understanding the dose- and radiation quality-dependence of the esophageal epithelium response to radiation may provide insights into the ability of radiation to promote ESCC. We have explored factors that may play a role in esophageal epithelial radiosensitivity and their potential relationship to ESCC risk. We have utilized a murine three-dimensional (3D) organoid model that recapitulates the morphology and functions of the stratified squamous epithelium of the esophagus to study persistent dose- and radiation quality-dependent changes. Interestingly, although high-linear energy transfer (LET) Fe ion exposure induced a more intense and persistent alteration of squamous differentiation and 53BP1 DNA damage foci levels as compared to Cs, the MAPK/SAPK stress pathway signaling showed similar altered levels for most phospho-proteins with both radiation qualities. In addition, the lower dose of high-LET exposure also revealed nearly the same degree of morphological changes, even though only ~36% of the cells were predicted to be hit at the lower 0.1 Gy dose, suggesting that a bystander effect may be induced. Although p38 and ERK/MAPK revealed the highest levels following high-LET exposure, the findings reveal that even a low dose (0.1 Gy) of both radiation qualities can elicit a persistent stress signaling response that may critically impact the differentiation gradient of the esophageal epithelium, providing novel insights into the pathogenesis of radiation-induced esophageal injury and early stage esophageal carcinogenesis.


Subject(s)
Epithelial Cells , Esophagus , Organoids , Animals , Organoids/radiation effects , Organoids/pathology , Mice , Esophagus/radiation effects , Esophagus/pathology , Epithelial Cells/radiation effects , Epithelial Cells/pathology , Epithelial Cells/metabolism , DNA Damage , Esophageal Squamous Cell Carcinoma/pathology , Linear Energy Transfer , Esophageal Neoplasms/pathology , Esophageal Neoplasms/metabolism , Cell Differentiation/radiation effects , Tumor Suppressor p53-Binding Protein 1/metabolism , MAP Kinase Signaling System/radiation effects , Radiation Tolerance
8.
Cells ; 13(10)2024 May 09.
Article in English | MEDLINE | ID: mdl-38786035

ABSTRACT

There are fewer investigations conducted on human primary endometrial epithelial cells (HPEECs) compared to human primary endometrial stromal cells (HPESCs). One of the main reasons is the scarcity of protocols enabling prolonged epithelial cell culture. Even though it is possible to culture HPEECs in 3D over a longer period of time, it is technically demanding. In this study, we successfully established a highly pure, stable, and long-term viable human conditionally reprogrammed endometrial epithelial cell line, designated as eCRC560. These cells stained positive for epithelial markers, estrogen and progesterone receptors, and epithelial cell-cell contacts but negative for stromal and endothelial cell markers. Estradiol (ES) reduced the abundance of ZO-1 in a time- and dose-dependent manner, in contrast to the dose-dependent increase with the progestin dienogest (DNG) when co-cultured with HPESCs. Moreover, ES significantly increased cell viability, cell migration, and invasion of the eCRC560 cells; all these effects were inhibited by pretreatment with DNG. DNG withdrawal led to a significantly disrupted monolayer of eCRC560 cells in co-culture with HPESCs, yet it markedly increased the adhesion of eCRC560 to the human mesothelial MeT-5A cells. The long-term viable eCRC560 cells are suitable for in vitro analysis of HPEECs to study the epithelial compartment of the human endometrium and endometrial pathologies.


Subject(s)
Cell Survival , Endometrium , Epithelial Cells , Estrogens , Progestins , Humans , Female , Endometrium/cytology , Endometrium/drug effects , Endometrium/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Progestins/pharmacology , Estrogens/pharmacology , Cell Survival/drug effects , Cell Movement/drug effects , Cell Line , Estradiol/pharmacology , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/cytology , Coculture Techniques , Time Factors , Cell Adhesion/drug effects
9.
Anim Biotechnol ; 35(1): 2344210, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38785376

ABSTRACT

The PPARGC1A gene plays a fundamental role in regulating cellular energy metabolism, including adaptive thermogenesis, mitochondrial biogenesis, adipogenesis, gluconeogenesis, and glucose/fatty acid metabolism. In a previous study, our group investigated seven SNPs in Mediterranean buffalo associated with milk production traits, and the current study builds on this research by exploring the regulatory influences of the PPARGC1A gene in buffalo mammary epithelial cells (BuMECs). Our findings revealed that knockdown of PPARGC1A gene expression significantly affected the growth of BuMECs, including proliferation, cell cycle, and apoptosis. Additionally, we observed downregulated triglyceride secretion after PPARGC1A knockdown. Furthermore, the critical genes related to milk production, including the STATS, BAD, P53, SREBF1, and XDH genes were upregulated after RNAi, while the FABP3 gene, was downregulated. Moreover, Silencing the PPARGC1A gene led to a significant downregulation of ß-casein synthesis in BuMECs. Our study provides evidence of the importance of the PPARGC1A gene in regulating cell growth, lipid, and protein metabolism in the buffalo mammary gland. In light of our previous research, the current study underscores the potential of this gene for improving milk production efficiency and overall dairy productivity in buffalo populations.


Subject(s)
Buffaloes , Epithelial Cells , Mammary Glands, Animal , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Animals , Buffaloes/genetics , Epithelial Cells/metabolism , Female , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/cytology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Milk , Gene Expression Regulation , Lactation/genetics , Cell Proliferation/genetics , Gene Knockdown Techniques , Apoptosis/genetics
10.
Commun Biol ; 7(1): 514, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38710749

ABSTRACT

Acute lung injury (ALI) is characterized by respiratory failure resulting from the disruption of the epithelial and endothelial barriers as well as immune system. In this study, we evaluated the therapeutic potential of airway epithelial cell-derived extracellular vesicles (EVs) in maintaining lung homeostasis. We isolated human bronchial epithelial cell-derived EVs (HBEC-EVs), which endogenously express various immune-related surface markers and investigated their immunomodulatory potential in ALI. In ALI cellular models, HBEC-EVs demonstrated immunosuppressive effects by reducing the secretion of proinflammatory cytokines in both THP-1 macrophages and HBECs. Mechanistically, these effects were partially ascribed to nine of the top 10 miRNAs enriched in HBEC-EVs, governing toll-like receptor-NF-κB signaling pathways. Proteomic analysis revealed the presence of proteins in HBEC-EVs involved in WNT and NF-κB signaling pathways, pivotal in inflammation regulation. ANXA1, a constituent of HBEC-EVs, interacts with formyl peptide receptor (FPR)2, eliciting anti-inflammatory responses by suppressing NF-κB signaling in inflamed epithelium, including type II alveolar epithelial cells. In a mouse model of ALI, intratracheal administration of HBEC-EVs reduced lung injury, inflammatory cell infiltration, and cytokine levels. Collectively, these findings suggest the therapeutic potential of HBEC-EVs, through their miRNAs and ANXA1 cargo, in mitigating lung injury and inflammation in ALI patients.


Subject(s)
Acute Lung Injury , Annexin A1 , Epithelial Cells , Extracellular Vesicles , Receptors, Formyl Peptide , Receptors, Lipoxin , Signal Transduction , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Humans , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Annexin A1/metabolism , Annexin A1/genetics , Animals , Mice , Receptors, Formyl Peptide/metabolism , Receptors, Formyl Peptide/genetics , Epithelial Cells/metabolism , Bronchi/metabolism , Bronchi/cytology , Male , Mice, Inbred C57BL , MicroRNAs/metabolism , MicroRNAs/genetics , NF-kappa B/metabolism , Cytokines/metabolism , THP-1 Cells
11.
Sci Rep ; 14(1): 10905, 2024 05 13.
Article in English | MEDLINE | ID: mdl-38740939

ABSTRACT

Cancer-derived cell lines are useful tools for studying cellular metabolism and xenobiotic toxicity, but they are not suitable for modeling the biological effects of food contaminants or natural biomolecules on healthy colonic epithelial cells in a normal genetic context. The toxicological properties of such compounds may rely on their oxidative properties. Therefore, it appears to be necessary to develop a dual-cell model in a normal genetic context that allows to define the importance of oxidative stress in the observed toxicity. Given that the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is considered to be the master regulator of antioxidant defenses, our aim was to develop a cellular model comparing normal and Nrf2-depleted isogenic cells to qualify oxidative stress-related toxicity. We generated these cells by using the CRISPR/Cas9 technique. Whole-genome sequencing enabled us to confirm that our cell lines were free of cancer-related mutations. We used 4-hydroxy-2-nonenal (HNE), a lipid peroxidation product closely related to oxidative stress, as a model molecule. Here we report significant differences between the two cell lines in glutathione levels, gene regulation, and cell viability after HNE treatment. The results support the ability of our dual-cell model to study the role of oxidative stress in xenobiotic toxicity.


Subject(s)
Epithelial Cells , NF-E2-Related Factor 2 , Oxidative Stress , Oxidative Stress/drug effects , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Animals , Mice , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Aldehydes/metabolism , Glutathione/metabolism , Cell Survival/drug effects , Cell Line , CRISPR-Cas Systems , Lipid Peroxidation/drug effects
12.
Front Immunol ; 15: 1362404, 2024.
Article in English | MEDLINE | ID: mdl-38745671

ABSTRACT

Introduction: The anti-inflammatory effect of green tea extract (GTE) has been confirmed in asthmatic mice, however, the pharmacological mechanism is not fully elucidated. Methods: To investigate the therapeutic efficacy of GTE in asthma and identify specific pathways, murine model of allergic asthma was established by ovalbumin (OVA) sensitization and the challenge for 4 weeks, with oral treatment using GTE and dexamethasone (DEX). Inflammatory cell counts, cytokines, OVA-specific IgE, airway hyperreactivity, and antioxidant markers in the lung were evaluated. Also, pulmonary histopathological analysis and western blotting were performed. In vitro, we established the model by stimulating the human airway epithelial cell line NCI-H292 using lipopolysaccharide, and treating with GTE and mitogen-activated protein kinases (MAPKs) inhibitors. Results: The GTE100 and GTE400 groups showed a decrease in airway hyperresponsiveness and the number of inflammatory cells in the bronchoalveolar lavage fluid (BALF) compared to the OVA group. GTE treatment also reduced interleukin (IL)-13, IL-5, and IL-4 levels in the BALF, and OVA-specific immunoglobulin E levels in the serum compared to those in the OVA group. GTE treatment decreased OVA-induced mucus secretion and airway inflammation. In addition, GTE suppressed the oxidative stress, and phosphorylation of MAPKs, which generally occurs after exposure to OVA. GTE administration also reduced matrix metalloproteinase-9 activity and protein levels. Conclusion: GTE effectively inhibited asthmatic respiratory inflammation and mucus hyperproduction induced by OVA inhalation. These results suggest that GTE has the potential to be used for the treatment of asthma.


Subject(s)
Asthma , Epithelial Cells , Matrix Metalloproteinase 9 , Oxidative Stress , Plant Extracts , Asthma/drug therapy , Asthma/immunology , Asthma/metabolism , Animals , Oxidative Stress/drug effects , Mice , Humans , Plant Extracts/pharmacology , Matrix Metalloproteinase 9/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Disease Models, Animal , Tea/chemistry , Female , Signal Transduction/drug effects , Mice, Inbred BALB C , Mitogen-Activated Protein Kinases/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/drug effects , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Cytokines/metabolism , Ovalbumin/immunology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
13.
Invest Ophthalmol Vis Sci ; 65(5): 24, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38748430

ABSTRACT

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.


Subject(s)
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
14.
PLoS One ; 19(5): e0300902, 2024.
Article in English | MEDLINE | ID: mdl-38748626

ABSTRACT

Tissue engineering predominantly relies on trial and error in vitro and ex vivo experiments to develop protocols and bioreactors to generate functional tissues. As an alternative, in silico methods have the potential to significantly reduce the timelines and costs of experimental programs for tissue engineering. In this paper, we propose a methodology to formulate, select, calibrate, and test mathematical models to predict cell population growth as a function of the biochemical environment and to design optimal experimental protocols for model inference of in silico model parameters. We systematically combine methods from the experimental design, mathematical statistics, and optimization literature to develop unique and explainable mathematical models for cell population dynamics. The proposed methodology is applied to the development of this first published model for a population of the airway-relevant bronchio-alveolar epithelial (BEAS-2B) cell line as a function of the concentration of metabolic-related biochemical substrates. The resulting model is a system of ordinary differential equations that predict the temporal dynamics of BEAS-2B cell populations as a function of the initial seeded cell population and the glucose, oxygen, and lactate concentrations in the growth media, using seven parameters rigorously inferred from optimally designed in vitro experiments.


Subject(s)
Cell Proliferation , Computer Simulation , Lung , Models, Biological , Humans , Cell Line , Lung/cytology , Lung/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Tissue Engineering/methods , Glucose/metabolism , Oxygen/metabolism
15.
Nat Commun ; 15(1): 3940, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750036

ABSTRACT

Hepatocytes play important roles in the liver, but in culture, they immediately lose function and dedifferentiate into progenitor-like cells. Although this unique feature is well-known, the dynamics and mechanisms of hepatocyte dedifferentiation and the differentiation potential of dedifferentiated hepatocytes (dediHeps) require further investigation. Here, we employ a culture system specifically established for hepatic progenitor cells to study hepatocyte dedifferentiation. We found that hepatocytes dedifferentiate with a hybrid epithelial/mesenchymal phenotype, which is required for the induction and maintenance of dediHeps, and exhibit Vimentin-dependent propagation, upon inhibition of the Hippo signaling pathway. The dediHeps re-differentiate into mature hepatocytes by forming aggregates, enabling reconstitution of hepatic tissues in vivo. Moreover, dediHeps have an unexpected differentiation potential into intestinal epithelial cells that can form organoids in three-dimensional culture and reconstitute colonic epithelia after transplantation. This remarkable plasticity will be useful in the study and treatment of intestinal metaplasia and related diseases in the liver.


Subject(s)
Cell Dedifferentiation , Cell Differentiation , Epithelial Cells , Hepatocytes , Animals , Hepatocytes/cytology , Hepatocytes/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Mice , Organoids/cytology , Organoids/metabolism , Epithelial-Mesenchymal Transition , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Cells, Cultured , Signal Transduction , Vimentin/metabolism , Hippo Signaling Pathway , Liver/cytology , Liver/metabolism , Mice, Inbred C57BL , Male , Cell Culture Techniques/methods
16.
J Transl Med ; 22(1): 458, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750454

ABSTRACT

BACKGROUND: Corneal injuries, often leading to severe vision loss or blindness, have traditionally been treated with the belief that limbal stem cells (LSCs) are essential for repair and homeostasis, while central corneal epithelial cells (CCECs) were thought incapable of such repair. However, our research reveals that CCECs can fully heal and maintain the homeostasis of injured corneas in rats, even without LSCs. We discovered that CXCL14, under PAX6's influence, significantly boosts the stemness, proliferation, and migration of CCECs, facilitating corneal wound healing and homeostasis. This finding introduces CXCL14 as a promising new drug target for corneal injury treatment. METHODS: To investigate the PAX6/CXCL14 regulatory axis's role in CCECs wound healing, we cultured human corneal epithelial cell lines with either increased or decreased expression of PAX6 and CXCL14 using adenovirus transfection in vitro. Techniques such as coimmunoprecipitation, chromatin immunoprecipitation, immunofluorescence staining, western blot, real-time PCR, cell colony formation, and cell cycle analysis were employed to validate the axis's function. In vivo, a rat corneal epithelial injury model was developed to further confirm the PAX6/CXCL14 axis's mechanism in repairing corneal damage and maintaining corneal homeostasis, as well as to assess the potential of CXCL14 protein as a therapeutic agent for corneal injuries. RESULTS: Our study reveals that CCECs naturally express high levels of CXCL14, which is significantly upregulated by PAX6 following corneal damage. We identified SDC1 as CXCL14's receptor, whose engagement activates the NF-κB pathway to stimulate corneal repair by enhancing the stemness, proliferative, and migratory capacities of CCECs. Moreover, our research underscores CXCL14's therapeutic promise for corneal injuries, showing that recombinant CXCL14 effectively accelerates corneal healing in rat models. CONCLUSION: CCECs play a critical and independent role in the repair of corneal injuries and the maintenance of corneal homeostasis, distinct from that of LSCs. The PAX6/CXCL14 regulatory axis is pivotal in this process. Additionally, our research demonstrates that the important function of CXCL14 in corneal repair endows it with the potential to be developed into a novel therapeutic agent for treating corneal injuries.


Subject(s)
Cell Proliferation , Chemokines, CXC , Corneal Injuries , Epithelium, Corneal , PAX6 Transcription Factor , Wound Healing , PAX6 Transcription Factor/metabolism , PAX6 Transcription Factor/genetics , Animals , Corneal Injuries/metabolism , Corneal Injuries/pathology , Humans , Chemokines, CXC/metabolism , Chemokines, CXC/genetics , Epithelium, Corneal/pathology , Epithelium, Corneal/metabolism , Rats, Sprague-Dawley , Epithelial Cells/metabolism , Rats , Cell Movement , Male , Cell Line
17.
Nat Commun ; 15(1): 3900, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724552

ABSTRACT

By incompletely understood mechanisms, type 2 (T2) inflammation present in the airways of severe asthmatics drives the formation of pathologic mucus which leads to airway mucus plugging. Here we investigate the molecular role and clinical significance of intelectin-1 (ITLN-1) in the development of pathologic airway mucus in asthma. Through analyses of human airway epithelial cells we find that ITLN1 gene expression is highly induced by interleukin-13 (IL-13) in a subset of metaplastic MUC5AC+ mucus secretory cells, and that ITLN-1 protein is a secreted component of IL-13-induced mucus. Additionally, we find ITLN-1 protein binds the C-terminus of the MUC5AC mucin and that its deletion in airway epithelial cells partially reverses IL-13-induced mucostasis. Through analysis of nasal airway epithelial brushings, we find that ITLN1 is highly expressed in T2-high asthmatics, when compared to T2-low children. Furthermore, we demonstrate that both ITLN-1 gene expression and protein levels are significantly reduced by a common genetic variant that is associated with protection from the formation of mucus plugs in T2-high asthma. This work identifies an important biomarker and targetable pathways for the treatment of mucus obstruction in asthma.


Subject(s)
Asthma , GPI-Linked Proteins , Interleukin-13 , Lectins , Mucin 5AC , Mucus , Child , Humans , Asthma/genetics , Asthma/metabolism , Cytokines , Epithelial Cells/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Interleukin-13/genetics , Interleukin-13/metabolism , Lectins/genetics , Lectins/metabolism , Mucin 5AC/genetics , Mucin 5AC/metabolism , Mucus/metabolism , Nasal Mucosa/metabolism , Polymorphism, Genetic , Respiratory Mucosa/metabolism
18.
J Nanobiotechnology ; 22(1): 236, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38724995

ABSTRACT

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.


Subject(s)
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
19.
Cells ; 13(9)2024 May 02.
Article in English | MEDLINE | ID: mdl-38727316

ABSTRACT

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.


Subject(s)
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
20.
Mol Biol Rep ; 51(1): 637, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38727927

ABSTRACT

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.


Subject(s)
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
SELECTION OF CITATIONS
SEARCH DETAIL
...