Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 19.710
1.
PLoS One ; 19(5): e0304365, 2024.
Article En | MEDLINE | ID: mdl-38820434

OBJECTIVE: To explore the molecular mechanism of Astragaloside IV (AS-IV) in alleviating renal fibrosis by inhibiting Urotensin II-induced pyroptosis and epithelial-mesenchymal transition of renal tubular epithelial cells. METHODS: Forty SD rats were randomly divided into control group without operation: gavage with 5ml/kg/d water for injection and UUO model group: gavage with 5ml/kg/d water for injection; UUO+ AS-IV group (gavage with AS-IV 20mg/kg/d; and UUO+ losartan potassium group (gavage with losartan potassium 10.3mg/kg/d, with 10 rats in each group. After 2 weeks, Kidney pathology, serum Urotensin II, and cAMP concentration were detected, and the expressions of NLRP3, GSDMD-N, Caspase-1, and IL-1ß were detected by immunohistochemistry. Rat renal tubular epithelial cells were cultured in vitro, and different concentrations of Urotensin II were used to intervene for 24h and 48h. Cell proliferation activity was detected using the CCK8 assay. Suitable concentrations of Urotensin II and intervention time were selected, and Urotensin II receptor antagonist (SB-611812), inhibitor of PKA(H-89), and AS-IV (15ug/ml) were simultaneously administered. After 24 hours, cells and cell supernatants from each group were collected. The cAMP concentration was detected using the ELISA kit, and the expression of PKA, α-SMA, FN, IL-1ß, NLRP3, GSDMD-N, and Caspase-1 was detected using cell immunofluorescence, Western blotting, and RT-PCR. RESULTS: Renal tissue of UUO rats showed renal interstitial infiltration, tubule dilation and atrophy, renal interstitial collagen fiber hyperplasia, and serum Urotensin II and cAMP concentrations were significantly higher than those in the sham operation group (p <0.05). AS-IV and losartan potassium intervention could alleviate renal pathological changes, and decrease serum Urotensin II, cAMP concentration levels, and the expressions of NLRP3, GSDMD-N, Caspase-1, and IL-1ß in renal tissues (p <0.05). Urotensin II at a concentration of 10-8 mol/L could lead to the decrease of cell proliferation, (p<0.05). Compared with the normal group, the cAMP level and the PKA expression were significantly increased (p<0.05). After intervention with AS-IV and Urotensin II receptor antagonist, the cAMP level and the expression of PKA were remarkably decreased (p<0.05). Compared with the normal group, the expression of IL-1ß, NLRP3, GSDMD-N, and Caspase-1 in the Urotensin II group was increased (p<0.05), which decreased in the AS-IV and H-89 groups. CONCLUSION: AS-IV can alleviate renal fibrosis by inhibiting Urotensin II-induced pyroptosis of renal tubular epithelial cells by regulating the cAMP/PKA signaling pathway.


Cyclic AMP-Dependent Protein Kinases , Cyclic AMP , Epithelial Cells , Fibrosis , Kidney Tubules , Pyroptosis , Rats, Sprague-Dawley , Saponins , Signal Transduction , Triterpenes , Urotensins , Animals , Saponins/pharmacology , Cyclic AMP/metabolism , Urotensins/metabolism , Rats , Cyclic AMP-Dependent Protein Kinases/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Epithelial Cells/pathology , Kidney Tubules/pathology , Kidney Tubules/metabolism , Kidney Tubules/drug effects , Triterpenes/pharmacology , Signal Transduction/drug effects , Pyroptosis/drug effects , Male , Epithelial-Mesenchymal Transition/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Kidney Diseases/metabolism , Kidney Diseases/drug therapy , Kidney Diseases/pathology , Kidney Diseases/etiology
2.
Article En | MEDLINE | ID: mdl-38780272

Sepsis-induced kidney injury (SAKI) has been frequently established as a prevailing complication of sepsis which is linked to unfavorable outcomes. Fatty acid-binding protein-4 (FABP4) has been proposed as a possible target for the treatment of SAKI. In the current work, we aimed to explore the role and underlying mechanism of FABP4 in lipopolysaccharide (LPS)-induced human renal tubular epithelial cell damage. In LPS-induced human kidney 2 (HK2) cells, FABP4 expression was tested by the reverse transcription-quantitative polymerase chain reaction and Western blot. Cell counting kit-8 method assayed cell viability. Inflammatory levels were detected using the enzyme-linked immunosorbent assay. Immunofluorescence staining measured the nuclear translocation of nuclear factor kappa B p65. Thiobarbituric acid-reactive substances assay and C11 BODIPY 581/591 probe were used to estimate the level of cellular lipid peroxidation. Fe2+ content was examined by the kit. In addition, the expression of proteins related to inflammation-, ferroptosis- and Janus kinase 2 (JAK2)/signal transducer, and activator of transcription 3 (STAT3) signaling was detected by the Western blot analysis. The results revealed that FABP4 was significantly upregulated in LPS-treated HK2 cells, the knockdown of which elevated the viability, whereas alleviated the inflammation and ferroptosis in HK2 cells challenged with LPS. In addition, down-regulation of FABP4 inactivated JAK2/STAT3 signaling. JAK2/STAT3 stimulator (colivelin) and ferroptosis activator (Erastin) partially restored the effects of FABP4 interference on LPS-triggered inflammation and ferroptosis in HK2 cells. Together, FABP4 knockdown inhibited ferroptosis to alleviate LPS-induced injury of renal tubular epithelial cells through suppressing JAK2/STAT3 signaling.


Epithelial Cells , Fatty Acid-Binding Proteins , Ferroptosis , Janus Kinase 2 , Kidney Tubules , Lipopolysaccharides , STAT3 Transcription Factor , Signal Transduction , Humans , Lipopolysaccharides/toxicity , Ferroptosis/drug effects , Janus Kinase 2/metabolism , Fatty Acid-Binding Proteins/metabolism , Fatty Acid-Binding Proteins/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Epithelial Cells/pathology , Signal Transduction/drug effects , Cell Line , Kidney Tubules/pathology , Kidney Tubules/metabolism , Kidney Tubules/drug effects , Acute Kidney Injury/metabolism , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Acute Kidney Injury/chemically induced
3.
Nat Med ; 30(5): 1349-1362, 2024 May.
Article En | MEDLINE | ID: mdl-38724705

Immune checkpoint inhibitor (ICI) therapy has revolutionized oncology, but treatments are limited by immune-related adverse events, including checkpoint inhibitor colitis (irColitis). Little is understood about the pathogenic mechanisms driving irColitis, which does not readily occur in model organisms, such as mice. To define molecular drivers of irColitis, we used single-cell multi-omics to profile approximately 300,000 cells from the colon mucosa and blood of 13 patients with cancer who developed irColitis (nine on anti-PD-1 or anti-CTLA-4 monotherapy and four on dual ICI therapy; most patients had skin or lung cancer), eight controls on ICI therapy and eight healthy controls. Patients with irColitis showed expanded mucosal Tregs, ITGAEHi CD8 tissue-resident memory T cells expressing CXCL13 and Th17 gene programs and recirculating ITGB2Hi CD8 T cells. Cytotoxic GNLYHi CD4 T cells, recirculating ITGB2Hi CD8 T cells and endothelial cells expressing hypoxia gene programs were further expanded in colitis associated with anti-PD-1/CTLA-4 therapy compared to anti-PD-1 therapy. Luminal epithelial cells in patients with irColitis expressed PCSK9, PD-L1 and interferon-induced signatures associated with apoptosis, increased cell turnover and malabsorption. Together, these data suggest roles for circulating T cells and epithelial-immune crosstalk critical to PD-1/CTLA-4-dependent tolerance and barrier function and identify potential therapeutic targets for irColitis.


Colitis , Immune Checkpoint Inhibitors , Intestinal Mucosa , Single-Cell Analysis , Humans , Immune Checkpoint Inhibitors/adverse effects , Colitis/chemically induced , Colitis/immunology , Colitis/genetics , Colitis/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/drug effects , Female , Male , Gene Expression Profiling , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Middle Aged , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Aged , Transcriptome , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Colon/pathology , Colon/immunology , Colon/drug effects , Epithelial Cells/immunology , Epithelial Cells/drug effects , Epithelial Cells/pathology
4.
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
5.
Sci Rep ; 14(1): 10905, 2024 05 13.
Article En | MEDLINE | ID: mdl-38740939

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.


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
6.
Front Immunol ; 15: 1362404, 2024.
Article En | MEDLINE | ID: mdl-38745671

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.


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
7.
Life Sci Alliance ; 7(7)2024 Jul.
Article En | MEDLINE | ID: mdl-38697845

Defective mitophagy in renal tubular epithelial cells is one of the main drivers of renal fibrosis in diabetic kidney disease. Our gene sequencing data showed the expression of PINK1 and BNIP3, two key molecules of mitophagy, was decreased in renal tissues of VDR-knockout mice. Herein, streptozotocin (STZ) was used to induce renal interstitial fibrosis in mice. VDR deficiency exacerbated STZ-induced renal impairment and defective mitophagy. Paricalcitol (pari, a VDR agonist) and the tubular epithelial cell-specific overexpression of VDR restored the expression of PINK1 and BNIP3 in the renal cortex and attenuated STZ-induced kidney fibrosis and mitochondrial dysfunction. In HK-2 cells under high glucose conditions, an increased level of α-SMA, COL1, and FN and a decreased expression of PINK1 and BNIP3 with severe mitochondrial damage were observed, and these alterations could be largely reversed by pari treatment. ChIP-qPCR and luciferase reporter assays showed VDR could positively regulate the transcription of Pink1 and Bnip3 genes. These findings reveal that VDR could restore mitophagy defects and attenuate STZ-induced fibrosis in diabetic mice through regulation of PINK1 and BNIP3.


Diabetes Mellitus, Experimental , Diabetic Nephropathies , Ergocalciferols , Membrane Proteins , Mice, Knockout , Mitophagy , Protein Kinases , Receptors, Calcitriol , Streptozocin , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/genetics , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Receptors, Calcitriol/metabolism , Receptors, Calcitriol/genetics , Mitophagy/genetics , Mitophagy/drug effects , Protein Kinases/metabolism , Protein Kinases/genetics , Humans , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/genetics , Male , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Fibrosis , Kidney Tubules/metabolism , Kidney Tubules/pathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Mice, Inbred C57BL , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Cell Line , Gene Expression Regulation/drug effects
8.
Int J Mol Sci ; 25(10)2024 May 13.
Article En | MEDLINE | ID: mdl-38791357

The lung is prone to infections from respiratory viruses such as Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). A challenge in combating these infections is the difficulty in targeting antiviral activity directly at the lung mucosal tract. Boosting the capability of the respiratory mucosa to trigger a potent immune response at the onset of infection could serve as a potential strategy for managing respiratory infections. This study focused on screening immunomodulators to enhance innate immune response in lung epithelial and immune cell models. Through testing various subfamilies and pathways of pattern recognition receptors (PRRs), the nucleotide-binding and oligomerization domain (NOD)-like receptor (NLR) family was found to selectively activate innate immunity in lung epithelial cells. Activation of NOD1 and dual NOD1/2 by the agonists TriDAP and M-TriDAP, respectively, increased the number of IL-8+ cells by engaging the NF-κB and interferon response pathways. Lung epithelial cells showed a stronger response to NOD1 and dual NOD1/2 agonists compared to control. Interestingly, a less-pronounced response to NOD1 agonists was noted in PBMCs, indicating a tissue-specific effect of NOD1 in lung epithelial cells without inducing widespread systemic activation. The specificity of the NOD agonist pathway was confirmed through gene silencing of NOD1 (siRNA) and selective NOD1 and dual NOD1/2 inhibitors in lung epithelial cells. Ultimately, activation induced by NOD1 and dual NOD1/2 agonists created an antiviral environment that hindered SARS-CoV-2 replication in vitro in lung epithelial cells.


COVID-19 , Epithelial Cells , Immunity, Innate , Lung , Nod1 Signaling Adaptor Protein , Nod2 Signaling Adaptor Protein , SARS-CoV-2 , Humans , Nod1 Signaling Adaptor Protein/metabolism , Nod1 Signaling Adaptor Protein/agonists , Immunity, Innate/drug effects , SARS-CoV-2/physiology , SARS-CoV-2/immunology , COVID-19/immunology , COVID-19/virology , Epithelial Cells/virology , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Epithelial Cells/immunology , Lung/immunology , Lung/virology , Lung/metabolism , Nod2 Signaling Adaptor Protein/agonists , Nod2 Signaling Adaptor Protein/metabolism , COVID-19 Drug Treatment , NF-kappa B/metabolism , Antiviral Agents/pharmacology , A549 Cells , Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/pharmacology , Signal Transduction/drug effects , Interleukin-8/metabolism
9.
Biochem Biophys Res Commun ; 717: 150061, 2024 Jul 12.
Article En | MEDLINE | ID: mdl-38718570

Epithelial mesenchymal transition (EMT) is a critical process implicated in the pathogenesis of retinal fibrosis and the exacerbation of diabetic retinopathy (DR) within retinal pigment epithelium (RPE) cells. Apigenin (AP), a potential dietary supplement for managing diabetes and its associated complications, has demonstrated inhibitory effects on EMT in various diseases. However, the specific impact and underlying mechanisms of AP on EMT in RPE cells remain poorly understood. In this study, we have successfully validated the inhibitory effects of AP on high glucose-induced EMT in ARPE-19 cells and diabetic db/db mice. Notably, our findings have identified CBP/p300 as a potential therapeutic target for EMT in RPE cells and have further substantiated that AP effectively downregulates the expression of EMT-related genes by attenuating the activity of CBP/p300, consequently reducing histone acetylation alterations within the promoter region of these genes. Taken together, our results provide novel evidence supporting the inhibitory effect of AP on EMT in RPE cells, and highlight the potential of specifically targeting CBP/p300 as a strategy for inhibiting retinal fibrosis in the context of DR.


Apigenin , Epithelial-Mesenchymal Transition , Glucose , Histones , Retinal Pigment Epithelium , Epithelial-Mesenchymal Transition/drug effects , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Animals , Apigenin/pharmacology , Acetylation/drug effects , Humans , Glucose/metabolism , Glucose/toxicity , Histones/metabolism , Cell Line , Mice , p300-CBP Transcription Factors/metabolism , p300-CBP Transcription Factors/antagonists & inhibitors , Mice, Inbred C57BL , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Diabetic Retinopathy/drug therapy , E1A-Associated p300 Protein/metabolism , Male , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , CREB-Binding Protein/metabolism , CREB-Binding Protein/genetics
10.
Cell Biol Toxicol ; 40(1): 33, 2024 May 21.
Article En | MEDLINE | ID: mdl-38769285

Fumonisin B1 (FB1), a water-soluble mycotoxin released by Fusarium moniliforme Sheld, is widely present in corn and its derivative products, and seriously endangers human life and health. Recent studies have reported that FB1 can lead to pyroptosis, however, the mechanisms by which FB1-induced pyroptosis remain indistinct. In the present study, we aim to investigate the mechanisms of pyroptosis in intestinal porcine epithelial cells (IPEC-J2) and the relationship between FB1-induced endoplasmic reticulum stress (ERS) and pyroptosis. Our experimental results showed that the pyroptosis protein indicators in IPEC-J2 were significantly increased after exposure to FB1. The ERS markers, including glucose-regulated Protein 78 (GRP78), PKR-like ER kinase protein (PERK), and preprotein translocation factor (Sec62) were also significantly increased. Using small interfering RNA silencing of PERK or Sec62, the results demonstrated that upregulation of Sec62 activates the PERK pathway, and activation of the PERK signaling pathway is upstream of FB1-induced pyroptosis. After using the ERS inhibitor 4-PBA reduced the FB1-triggered intestinal injury by the Sec62-PERK pathway. In conclusion, we found that FB1 induced pyroptosis by upregulating Sec62 to activate the PERK pathway, and mild ERS alleviates FB1-triggered damage. It all boils down to one fact, the study provides a new perspective for further, and improving the toxicological mechanism of FB1.


Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Pyroptosis , Signal Transduction , eIF-2 Kinase , Pyroptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Animals , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , Swine , Signal Transduction/drug effects , Endoplasmic Reticulum Chaperone BiP/metabolism , Cell Line , Intestines/drug effects , Intestines/pathology , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Fumonisins
11.
Ecotoxicol Environ Saf ; 278: 116456, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38744067

Long non-coding RNAs (LncRNAs) are dysregulated in a variety of human diseases and are highly involved in the development and progression of tumors. Studies on lncRNAs associated with cow mastitis have been lagging behind compared to humans or model animals, therefore, the aim of this study was to explore the mechanism of LncRNAs (CMR) involved in autoprotection against S. aureus mastitis in Bovine Mammary Epithelial Cells (BMECs). First, qRT-PCR was used to examine the relative expression of CMR in a S. aureus mastitis model of BMECs. Then, cell proliferation and apoptosis were detected by EdU and apoptosis assay. Finally, the targeting relationship between miRNAs and mRNA/LncRNAs was determined by dual luciferase reporter gene, qRT-PCR and western blotting techniques. The results showed that CMR was upregulated in the S. aureus mastitis model of BMECs and promoted the expression of inflammatory factors, and SiRNA-mediated CMR inhibited the proliferation of mammary epithelial cells and induced apoptosis. Mechanistically, CMR acts as a competitive endogenous RNA (ceRNA) sponge miR-877, leading to upregulation of FOXM1, a target of miR-877. Importantly, either miR-877 overexpression or FOXM1 inhibition abrogated CMR knockdown-induced apoptosis promoting cell proliferation and reducing inflammatory factor expression levels. In summary, CMR is involved in the regulation of autoprotection against S. aureus mastitis through the miR-877/FOXM1 axis in BMECs and induces immune responses in mammary tissues and cells of dairy cows, providing an important reference for subsequent prevention and control of cow mastitis and the development of targeted drugs.


Mastitis, Bovine , MicroRNAs , RNA, Long Noncoding , Staphylococcus aureus , Animals , Cattle , RNA, Long Noncoding/genetics , MicroRNAs/genetics , Female , Mastitis, Bovine/genetics , Mastitis, Bovine/microbiology , Apoptosis , Forkhead Box Protein M1/genetics , Cell Proliferation , Epithelial Cells/drug effects , Staphylococcal Infections/genetics
12.
Environ Health Perspect ; 132(5): 57006, 2024 May.
Article En | MEDLINE | ID: mdl-38771937

BACKGROUND: Uranium exposure remains an important environmental legacy and physiological health concern, with hundreds of abandoned uranium mines located in the Southwestern United States largely impacting underserved indigenous communities. The negative effects of heavy metals on barrier permeability and inhibition of intestinal epithelial healing have been described; however, transcriptomic changes within the intestinal epithelial cells and impacts on lineage differentiation are largely unknown. OBJECTIVES: Herein, we sought to determine the molecular and cellular changes that occur in the colon in response to uranium bearing dust (UBD) exposure. METHODS: Human colonoids from three biologically distinct donors were acutely exposed to UBD then digested for single cell RNA sequencing to define the molecular changes that occur to specific identities of colonic epithelial cells. Validation in colonoids was assessed using morphological and imaging techniques. RESULTS: Human colonoids acutely exposed to UBD exhibited disrupted proliferation and hyperplastic differentiation of the secretory lineage cell, enteroendocrine cells (EEC). Single-cell RNA sequencing also showed more EEC subtypes present in UBD-exposed colonoids. DISCUSSION: These findings highlight the significance of crypt-based proliferative cells and secretory cell differentiation using human colonoids to model major colonic responses to uranium-bearing particulate dust exposure. https://doi.org/10.1289/EHP13855.


Colon , Dust , Single-Cell Analysis , Uranium , Humans , Uranium/toxicity , Colon/drug effects , Epithelial Cells/drug effects
13.
Cells ; 13(10)2024 May 09.
Article En | MEDLINE | ID: mdl-38786035

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.


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
14.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(5): 411-418, 2024 May.
Article Zh | MEDLINE | ID: mdl-38790097

Objective To explore the effects of aloperine (Alo) on cigarette smoke-induced injury in human bronchial epithelial cells and its potential mechanism. Methods After human bronchial epithelial 16HBE cells were co-treated by 100 mL/L cigarette smoke extract (CSE) and various concentrations (50,100 and 200 µmol/L) of Alo, cell viability was assessed using CCK-8 assay. Lactate dehydrogenase (LDH) activity was measured with a related kit. Cell apoptosis was evaluated using the terminal-deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay (TUNEL) and Western blot analysis. The levels of inflammatory factors were detected by ELISA. Oxidative stress levels were assessed using 2'7'-dichlorofluorescin diacetate (DCFH-DA) staining. The expression of Toll-like receptor 4 (TLR4)/nuclear factor-kappaB (NF-κB)/NLR family pyrin domain containing 3 (NLRP3) signaling-associated proteins was measured by Western blot analysis. After cells were co-treated with 100 mL/L CSE and 200 µmol/L Alo, the aforementioned assays were applied to evaluate the effects of TLR4 overexpression on the TLR4/NF-κB/NLRP3 signaling, LDH activity, apoptosis, inflammatory response and oxidative stress in cells. Results CSE exposure might inhibit 16HBE cell viability, increase LDH activity, apoptosis, inflammatory response and oxidative stress levels and activate TLR4/NF-κB/NLRP3 signaling. Treatment with Alo promoted cell viability, decreased LDH activity, cell apoptosis, inflammation and oxidative stress levels, and inactivated TLR4/NF-κB/NLRP3 signaling. Furthermore, TLR4 overexpression might reverse the protective role of Alo treatment in CSE-induced injury in 16HBE cells. Conclusion Alo may ameliorate CSE-induced injury in human bronchial epithelial cells via inhibiting TLR4/NF-κB/NLRP3 signaling.


Apoptosis , Bronchi , Epithelial Cells , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Quinolizidines , Signal Transduction , Toll-Like Receptor 4 , Humans , Toll-Like Receptor 4/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Bronchi/cytology , Bronchi/metabolism , Bronchi/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Apoptosis/drug effects , Quinolizidines/pharmacology , Smoke/adverse effects , Oxidative Stress/drug effects , Cell Survival/drug effects , Cell Line , Nicotiana/adverse effects
15.
Respir Res ; 25(1): 212, 2024 May 18.
Article En | MEDLINE | ID: mdl-38762455

Paraquat (PQ) is a widely used herbicide and a common cause of poisoning that leads to pulmonary fibrosis with a high mortality rate. However, the underlying mechanisms of PQ-induced pulmonary fibrosis and whether pulmonary epithelial cell senescence is involved in the process remain elusive. In this study, PQ-induced pulmonary epithelial cell senescence and Hippo-YAP/TAZ activation were observed in both C57BL/6 mice and human epithelial cells. PQ-induced senescent pulmonary epithelial cells promoted lung fibroblast transformation through secreting senescence-associated secretory phenotype (SASP) factors. Yap/Taz knockdown in mice lungs significantly decreased the expression of downstream profibrotic protein Ctgf and senescent markers p16 and p21, and alleviated PQ-induced pulmonary fibrosis. Interfering YAP/TAZ in senescent human pulmonary epithelial cells resulted in decreased expression of the anti-apoptosis protein survivin and elevated level of apoptosis. In conclusion, our findings reveal a novel mechanism by which the involvement of Hippo-YAP/TAZ activation in pulmonary epithelial cell senescence mediates the pathogenesis of PQ-induced pulmonary fibrosis, thereby offering novel insights and potential targets for the clinical management of PQ poisoning as well as providing the mechanistic insight of the involvement of Yap/Taz activation in cell senescence in pulmonary fibrosis and its related pulmonary disorders. The YIN YANG balance between cell senescence and apoptosis is important to maintain the homeostasis of the lung, the disruption of which will lead to disease.


Adaptor Proteins, Signal Transducing , Cellular Senescence , Mice, Inbred C57BL , Paraquat , Pulmonary Fibrosis , Transcription Factors , YAP-Signaling Proteins , Animals , Cellular Senescence/drug effects , Cellular Senescence/physiology , YAP-Signaling Proteins/metabolism , Humans , Mice , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Adaptor Proteins, Signal Transducing/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Paraquat/toxicity , Male , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Epithelial Cells/pathology , Trans-Activators/metabolism , Trans-Activators/genetics
16.
J Agric Food Chem ; 72(20): 11733-11745, 2024 May 22.
Article En | MEDLINE | ID: mdl-38725145

Amino acids are essential for the activation of the mechanistic target of rapamycin (mTOR), but the corresponding molecular mechanism is not yet fully understood. We previously found that Met stimulated eukaryotic elongation factor α (eEF1Bα) nuclear localization in bovine mammary epithelial cells (MECs). Herein, we explored the role and molecular mechanism of eEF1Bα in methionine (Met)- and leucine (Leu)-stimulated mTOR gene transcription and milk synthesis in MECs. eEF1Bα knockdown decreased milk protein and fat synthesis, cell proliferation, and mTOR mRNA expression and phosphorylation, whereas eEF1Bα overexpression had the opposite effects. QE-MS analysis detected that eEF1Bα was phosphorylated at Ser106 in the nucleus and Met and Leu stimulated p-eEF1Bα nuclear localization. eEF1Bα knockdown abrogated the stimulation of Met and Leu by mTOR mRNA expression and phosphorylation, and this regulatory role was dependent on its phosphorylation. Akt knockdown blocked the stimulation of Met and Leu by eEF1Bα and p-eEF1Bα expression. ChIP-PCR detected that p-eEF1Bα bound only to the -548 to -793 nt site in the mTOR promoter, and ChIP-qPCR further detected that Met and Leu stimulated this binding. eEF1Bα mediated Met and Leu' stimulation on mTOR mRNA expression and phosphorylation through inducing AT-rich interaction domain 1A (ARID1A) ubiquitination degradation, and this process depended on eEF1Bα phosphorylation. p-eEF1Bα interacted with ARID1A and ubiquitin protein ligase E3 module N-recognition 5 (UBR5), and UBR5 knockdown rescued the decrease of the ARID1A protein level by eEF1Bα overexpression. Both eEF1Bα and p-eEF1Bα were highly expressed in mouse mammary gland tissues during the lactating period. In summary, we reveal that Met and Leu stimulate mTOR transcriptional activation and milk protein and fat synthesis in MECs through eEF1Bα-UBR5-ARID1A signaling.


Epithelial Cells , Leucine , Mammary Glands, Animal , Methionine , Milk , Signal Transduction , TOR Serine-Threonine Kinases , Animals , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Cattle , Female , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Signal Transduction/drug effects , Methionine/metabolism , Methionine/pharmacology , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/cytology , Milk/chemistry , Milk/metabolism , Leucine/pharmacology , Leucine/metabolism , Mice , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Peptide Elongation Factor 1/genetics , Peptide Elongation Factor 1/metabolism
17.
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
18.
Int Immunopharmacol ; 134: 112165, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38692017

Particulate matter (PM) is considered the fundamental component of atmospheric pollutants and is associated with the pathogenesis of many respiratory diseases. Fibroblast growth factor 10 (FGF10) mediates mesenchymal-epithelial signaling and has been linked with the repair process of PM-induced lung injury (PMLI). However, the pathogenic mechanism of PMLI and the specific FGF10 protective mechanism against this injury are still undetermined. PM was administered in vivo into murine airways or in vitro to human bronchial epithelial cells (HBECs), and the inflammatory response and ferroptosis-related proteins SLC7A11 and GPX4 were assessed. The present research investigates the FGF10-mediated regulation of ferroptosis in PMLI mice models in vivo and HBECs in vitro. The results showed that FGF10 pretreatment reduced PM-mediated oxidative damage and ferroptosis in vivo and in vitro. Furthermore, FGF10 pretreatment led to reduced oxidative stress, decreased secretion of inflammatory mediators, and activation of the Nrf2-dependent antioxidant signaling. Additionally, silencing of Nrf2 using siRNA in the context of FGF10 treatment attenuated the effect on ferroptosis. Altogether, both in vivo and in vitro assessments confirmed that FGF10 protects against PMLI by inhibiting ferroptosis via the Nrf2 signaling. Thus, FGF10 can be used as a novel ferroptosis suppressor and a potential treatment target in PMLI.


Ferroptosis , Fibroblast Growth Factor 10 , Lung Injury , Mice, Inbred C57BL , NF-E2-Related Factor 2 , Oxidative Stress , Particulate Matter , Signal Transduction , Ferroptosis/drug effects , NF-E2-Related Factor 2/metabolism , Animals , Particulate Matter/toxicity , Humans , Signal Transduction/drug effects , Fibroblast Growth Factor 10/metabolism , Fibroblast Growth Factor 10/genetics , Mice , Oxidative Stress/drug effects , Lung Injury/chemically induced , Lung Injury/metabolism , Lung Injury/pathology , Lung Injury/prevention & control , Male , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Cell Line , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Disease Models, Animal , Amino Acid Transport System y+
19.
Biomed Pharmacother ; 175: 116674, 2024 Jun.
Article En | MEDLINE | ID: mdl-38703509

Numerous cases of lung injury caused by viral infection were reported during the coronavirus disease-19 pandemic. While there have been significant efforts to develop drugs that block viral infection and spread, the development of drugs to reduce or reverse lung injury has been a lower priority. This study aimed to identify compounds from a library of compounds that prevent viral infection that could reduce and prevent lung epithelial cell damage. We investigated the cytotoxicity of the compounds, their activity in inhibiting viral spike protein binding to cells, and their activity in reducing IL-8 production in lung epithelial cells damaged by amodiaquine (AQ). We identified N-(4-(4-methoxyphenoxy)-3-methylphenyl)-N-methylacetamide (MPoMA) as a non-cytotoxic inhibitor against viral infection and AQ-induced cell damage. MPoMA inhibited the expression of IL-8, IL-6, IL-1ß, and fibronectin induced by AQ and protected against AQ-induced morphological changes. However, MPoMA did not affect basal IL-8 expression in lung epithelial cells in the absence of AQ. Further mechanistic analysis confirmed that MPoMA selectively promoted the proteasomal degradation of inflammatory mediator p65, thereby reducing intracellular p65 expression and p65-mediated inflammatory responses. MPoMA exerted potent anti-inflammatory and protective functions in epithelial cells against LPS-induced acute lung injury in vivo. These findings suggest that MPoMA may have beneficial effects in suppressing viral infection and preventing lung epithelial cell damage through the degradation of p65 and inhibition of the production of inflammatory cytokines.


Epithelial Cells , Animals , Humans , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Mice , Lung/pathology , Lung/drug effects , Lung/metabolism , Transcription Factor RelA/metabolism , COVID-19 Drug Treatment , A549 Cells , SARS-CoV-2/drug effects , COVID-19/prevention & control , Proteolysis/drug effects , Lung Injury/prevention & control , Lung Injury/pathology , Lung Injury/metabolism , Lung Injury/virology , Male , Acute Lung Injury/prevention & control , Acute Lung Injury/pathology , Acute Lung Injury/metabolism , Acetamides/pharmacology
20.
Biomaterials ; 309: 122593, 2024 Sep.
Article En | MEDLINE | ID: mdl-38713971

Posterior capsule opacification (PCO) is a predominant postoperative complication, often leading to visual impairment due to the aberrant proliferation and adhesion of lens epithelial cells (LECs) and protein precipitates subsequent to intraocular lens (IOL) implantation. To address this clinical issue, a foldable and antifouling sharp-edged IOL implant based on naturally-derived cellulose hydrogel is synthesized. The mechanical strength and transparency of the hydrogel is enhanced via repeated freeze-thaw (FT) cycles. The incorporated zwitterionic modifications can remarkably prevent the incidence of PCO by exhibiting proteins repulsion and cell anti-adhesion properties. The graft of dopamine onto both the haptic and the periphery of the posterior surface ensures the adhesion of the hydrogel to the posterior capsule and impedes the migration of LECs without compromising transparency. In in vivo study, the zwitterionic modified foldable hydrogel exhibits uveal and capsular biocompatibility synchronously with no signs of inflammatory response and prevent PCO formation, better than that of commercialized and PEG-modified IOL. With foldability, endurability, antifouling effect, and adhesive to posterior capsule, the reported hydrogel featuring heterogeneous surface design displays great potential to eradicate PCO and attain post-operative efficacy after cataract surgery.


Capsule Opacification , Lenses, Intraocular , Capsule Opacification/prevention & control , Animals , Hydrogels/chemistry , Rabbits , Humans , Freezing , Epithelial Cells/drug effects , Biocompatible Materials/chemistry
...