Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 15.063
Filter
1.
Zhonghua Bing Li Xue Za Zhi ; 53(6): 541-545, 2024 Jun 08.
Article in Chinese | MEDLINE | ID: mdl-38825897

ABSTRACT

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.


Subject(s)
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.
Sci Rep ; 14(1): 12879, 2024 06 05.
Article in English | MEDLINE | ID: mdl-38839896

ABSTRACT

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.


Subject(s)
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
4.
Article in English | MEDLINE | ID: mdl-38780272

ABSTRACT

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.


Subject(s)
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
5.
Nat Med ; 30(5): 1349-1362, 2024 May.
Article in English | MEDLINE | ID: mdl-38724705

ABSTRACT

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.


Subject(s)
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
6.
Int J Mol Sci ; 25(9)2024 May 02.
Article in English | MEDLINE | ID: mdl-38732189

ABSTRACT

Sjögren's Disease (SjD) is an autoimmune disease of the exocrine tissues. Etiological events result in the loss of epithelial homeostasis alongside extracellular matrix (ECM) destruction within the salivary and lacrimal glands, followed by immune cell infiltration. In this review, we have assessed the current understanding of epithelial-mesenchymal transition (EMT)-associated changes within the salivary epithelium potentially involved in salivary dysfunction and SjD pathogenesis. We performed a PubMed literature review pertaining to the determination of pathogenic events that lead to EMT-related epithelial dysfunction and signaling in SjD. Molecular patterns of epithelial dysfunction in SjD salivary glands share commonalities with EMT mediating wound healing. Pathological changes altering salivary gland integrity and function may precede direct immune involvement while perpetuating MMP9-mediated ECM destruction, inflammatory mediator expression, and eventual immune cell infiltration. Dysregulation of EMT-associated factors is present in the salivary epithelium of SjD and may be significant in initiating and perpetuating the disease. In this review, we further highlight the gap regarding mechanisms that drive epithelial dysfunction in salivary glands in the early or subclinical pre-lymphocytic infiltration stages of SjD.


Subject(s)
Epithelial-Mesenchymal Transition , Salivary Glands , Sjogren's Syndrome , Humans , Sjogren's Syndrome/pathology , Sjogren's Syndrome/metabolism , Salivary Glands/pathology , Salivary Glands/metabolism , Animals , Epithelium/pathology , Epithelium/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Signal Transduction , Extracellular Matrix/metabolism
7.
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
8.
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
9.
Toxicol Appl Pharmacol ; 487: 116954, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38705402

ABSTRACT

Dual-specificity phosphatase 26 (DUSP26) acts as a pivotal player in the transduction of signalling cascades with its dephosphorylating activity. Currently, DUSP26 attracts extensive attention due to its particular function in several pathological conditions. However, whether DUSP26 plays a role in kidney ischaemia-reperfusion (IR) injury is unknown. Aims of the current work were to explore the relevance of DUSP26 in kidney IR damage. DUSP26 levels were found to be decreased in renal tubular epithelial cells following hypoxia-reoxygenation (HR) and kidney samples subjected to IR treatments. DUSP26-overexpressed renal tubular epithelial cells exhibited protection against HR-caused apoptosis and inflammation, while DUSP26-depleted renal tubular epithelial cells were more sensitive to HR damage. Upregulation of DUSP26 in rat kidneys by infecting adenovirus expressing DUSP26 markedly ameliorated kidney injury caused by IR, while also effectively reducing apoptosis and inflammation. The mechanistic studies showed that the activation of transforming growth factor-ß-activated kinase 1 (TAK1)-JNK/p38 MAPK, contributing to kidney injury under HR or IR conditions, was restrained by increasing DUSP26 expression. Pharmacological restraint of TAK1 markedly diminished DUSP26-depletion-exacebated effects on JNK/p38 activation and HR injury of renal tubular cells. The work reported a renal-protective function of DUSP26, which protects against IR-related kidney damage via the intervention effects on the TAK1-JNK/p38 axis. The findings laid a foundation for understanding the molecular pathogenesis of kidney IR injury and provide a prospective target for treating this condition.


Subject(s)
Apoptosis , Epithelial Cells , Kidney Tubules , MAP Kinase Kinase Kinases , Rats, Sprague-Dawley , Reperfusion Injury , p38 Mitogen-Activated Protein Kinases , Animals , Reperfusion Injury/pathology , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Male , Kidney Tubules/pathology , Kidney Tubules/metabolism , Rats , p38 Mitogen-Activated Protein Kinases/metabolism , Dual-Specificity Phosphatases/metabolism , Dual-Specificity Phosphatases/genetics , Cell Line , Acute Kidney Injury/pathology , Acute Kidney Injury/metabolism , Inflammation/pathology , Inflammation/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Mitogen-Activated Protein Kinase Phosphatases/genetics , Signal Transduction/physiology
10.
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
11.
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
12.
Sci Rep ; 14(1): 12044, 2024 05 27.
Article in English | MEDLINE | ID: mdl-38802480

ABSTRACT

This study tackles the persistent prognostic and management challenges of clear cell renal cell carcinoma (ccRCC), despite advancements in multimodal therapies. Focusing on anoikis, a critical form of programmed cell death in tumor progression and metastasis, we investigated its resistance in cancer evolution. Using single-cell RNA sequencing from seven ccRCC patients, we assessed the impact of anoikis-related genes (ARGs) and identified differentially expressed genes (DEGs) in Anoikis-related epithelial subclusters (ARESs). Additionally, six ccRCC RNA microarray datasets from the GEO database were analyzed for robust DEGs. A novel risk prognostic model was developed through LASSO and multivariate Cox regression, validated using BEST, ULCAN, and RT-PCR. The study included functional enrichment, immune infiltration analysis in the tumor microenvironment (TME), and drug sensitivity assessments, leading to a predictive nomogram integrating clinical parameters. Results highlighted dynamic ARG expression patterns and enhanced intercellular interactions in ARESs, with significant KEGG pathway enrichment in MYC + Epithelial subclusters indicating enhanced anoikis resistance. Additionally, all ARESs were identified in the spatial context, and their locational relationships were explored. Three key prognostic genes-TIMP1, PECAM1, and CDKN1A-were identified, with the high-risk group showing greater immune infiltration and anoikis resistance, linked to poorer prognosis. This study offers a novel ccRCC risk signature, providing innovative approaches for patient management, prognosis, and personalized treatment.


Subject(s)
Anoikis , Biomarkers, Tumor , Carcinoma, Renal Cell , Gene Expression Regulation, Neoplastic , Kidney Neoplasms , Tumor Microenvironment , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Anoikis/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/metabolism , Prognosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Tumor Microenvironment/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Male , Female , Gene Expression Profiling , Nomograms
13.
Biochem Biophys Res Commun ; 717: 150061, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38718570

ABSTRACT

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.


Subject(s)
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
14.
Biomed Pharmacother ; 175: 116674, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703509

ABSTRACT

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.


Subject(s)
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
15.
PLoS One ; 19(5): e0304365, 2024.
Article in English | MEDLINE | ID: mdl-38820434

ABSTRACT

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.


Subject(s)
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
16.
J Histochem Cytochem ; 72(5): 289-307, 2024 05.
Article in English | MEDLINE | ID: mdl-38725414

ABSTRACT

Several types of cytotoxic insults disrupt endoplasmic reticulum (ER) homeostasis, cause ER stress, and activate the unfolded protein response (UPR). The role of ER stress and UPR activation in hypersensitivity pneumonitis (HP) has not been described. HP is an immune-mediated interstitial lung disease that develops following repeated inhalation of various antigens in susceptible and sensitized individuals. The aim of this study was to investigate the lung expression and localization of the key effectors of the UPR, BiP/GRP78, CHOP, and sXBP1 in HP patients compared with control subjects. Furthermore, we developed a mouse model of HP to determine whether ER stress and UPR pathway are induced during this pathogenesis. In human control lungs, we observed weak positive staining for BiP in some epithelial cells and macrophages, while sXBP1 and CHOP were negative. Conversely, strong BiP, sXBP1- and CHOP-positive alveolar and bronchial epithelial, and inflammatory cells were identified in HP lungs. We also found apoptosis and autophagy markers colocalization with UPR proteins in HP lungs. Similar results were obtained in lungs from an HP mouse model. Our findings suggest that the UPR pathway is associated with the pathogenesis of HP.


Subject(s)
Alveolitis, Extrinsic Allergic , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Epithelial Cells , Heat-Shock Proteins , Transcription Factor CHOP , Unfolded Protein Response , X-Box Binding Protein 1 , Animals , Alveolitis, Extrinsic Allergic/pathology , Alveolitis, Extrinsic Allergic/immunology , Alveolitis, Extrinsic Allergic/metabolism , Humans , Mice , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics , Heat-Shock Proteins/metabolism , Transcription Factor CHOP/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Male , Lung/pathology , Lung/immunology , Lung/metabolism , DNA-Binding Proteins/metabolism , Regulatory Factor X Transcription Factors/metabolism , Transcription Factors/metabolism , Disease Models, Animal , Middle Aged , Mice, Inbred C57BL , Adult , Inflammation/pathology , Inflammation/metabolism , Inflammation/immunology
17.
Nature ; 629(8013): 869-877, 2024 May.
Article in English | MEDLINE | ID: mdl-38693267

ABSTRACT

Airway hillocks are stratified epithelial structures of unknown function1. Hillocks persist for months and have a unique population of basal stem cells that express genes associated with barrier function and cell adhesion. Hillock basal stem cells continually replenish overlying squamous barrier cells. They exhibit dramatically higher turnover than the abundant, largely quiescent classic pseudostratified airway epithelium. Hillocks resist a remarkably broad spectrum of injuries, including toxins, infection, acid and physical injury because hillock squamous cells shield underlying hillock basal stem cells from injury. Hillock basal stem cells are capable of massive clonal expansion that is sufficient to resurface denuded airway, and eventually regenerate normal airway epithelium with each of its six component cell types. Hillock basal stem cells preferentially stratify and keratinize in the setting of retinoic acid signalling inhibition, a known cause of squamous metaplasia2,3. Here we show that mouse hillock expansion is the cause of vitamin A deficiency-induced squamous metaplasia. Finally, we identify human hillocks whose basal stem cells generate functional squamous barrier structures in culture. The existence of hillocks reframes our understanding of airway epithelial regeneration. Furthermore, we show that hillocks are one origin of 'squamous metaplasia', which is long thought to be a precursor of lung cancer.


Subject(s)
Cell Plasticity , Epithelial Cells , Regeneration , Respiratory Mucosa , Stem Cells , Animals , Female , Humans , Male , Mice , Epithelial Cells/cytology , Epithelial Cells/pathology , Metaplasia/etiology , Metaplasia/pathology , Respiratory Mucosa/cytology , Respiratory Mucosa/injuries , Respiratory Mucosa/pathology , Stem Cells/cytology , Tretinoin/metabolism , Tretinoin/pharmacology , Vitamin A/metabolism , Vitamin A/pharmacology , Lung Neoplasms/etiology , Lung Neoplasms/pathology , Mice, Inbred C57BL
18.
Eur J Pharmacol ; 975: 176640, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38750716

ABSTRACT

Anti-partial epithelial-mesenchymal transition (pEMT) treatment of renal tubular epithelial cells (TECs) represents a promising therapeutic approach. Hyperuricemia nephropathy (HN) arises as a consequence of hyperuricemia (HUA)-induced tubulointerstitial fibrosis (TIF). Studies have suggested that the Ras homolog member A (RhoA)/Rho-associated kinase (ROCK) pathway is a crucial signaling transduction system in renal fibrosis. Fasudil, a RhoA/ROCK inhibitor, has exhibited the potential to prevent fibrosis progress. However, its impact on the pEMT of TECs in HN remains unclear. Here, an HN rat model and an uric acid (UA)-stimulated human kidney 2 (HK2) cell model were established and treated with Fasudil to explore its effects. Furthermore, the underlying mechanism of action involved in the attenuation of pEMT in TECs by Fasudil during HN was probed by using multiple molecular approaches. The HN rat model exhibited significant renal dysfunction and histopathological damage, whereas in vitro and in vivo experiments further confirmed the pEMT status accompanied by RhoA/ROCK pathway activation and oxidative stress in tubular cells exposed to UA. Notably, Fasudil ameliorated these pathological changes, and this was consistent with the trend of ROCK silencing in vitro. Mechanistically, we identified the Neh2 domain of nuclear factor erythroid 2-related factor 2 (Nrf2) as a target of Fasudil for the first time. Fasudil targets Nrf2 activation and antagonizes oxidative stress to attenuate the pEMT of TECs in HN. Our findings suggest that Fasudil attenuates oxidative stress-induced pEMT of TECs in HN by targeting Nrf2 activation. Thus, Fasudil is a potential therapeutic agent for the treatment of HN.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine , Epithelial Cells , Epithelial-Mesenchymal Transition , Hyperuricemia , Kidney Diseases , Kidney Tubules , NF-E2-Related Factor 2 , Oxidative Stress , NF-E2-Related Factor 2/metabolism , Animals , Epithelial-Mesenchymal Transition/drug effects , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/therapeutic use , Oxidative Stress/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Hyperuricemia/drug therapy , Hyperuricemia/metabolism , Humans , Rats , Male , Kidney Tubules/drug effects , Kidney Tubules/pathology , Kidney Tubules/metabolism , Cell Line , Kidney Diseases/drug therapy , Kidney Diseases/pathology , Kidney Diseases/metabolism , rho-Associated Kinases/metabolism , rho-Associated Kinases/antagonists & inhibitors , Rats, Sprague-Dawley , Disease Models, Animal , Signal Transduction/drug effects
19.
Toxicology ; 505: 153827, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729513

ABSTRACT

Pyroptosis is a form of programmed cell death characterized by gasdermin (GSDM)-mediated pore formation in the cell membrane, resulting in the release of pro-inflammatory cytokines and cellular lysis. Increasing evidence has shown that pyroptosis is responsible for the progression of various pulmonary disorders. The inhalation of polyhexamethylene guanidine (PHMG) causes severe lung inflammation and pulmonary toxicity; however, the underlying mechanisms are unknown. Therefore, in this study, we investigate the role of pyroptosis in PHMG-induced pulmonary toxicity. We exposed bronchial epithelial cells, BEAS-2B, to PHMG phosphate (PHMG-p) and evaluated cell death type, reactive oxygen species (ROS) levels, and relative expression levels of pyroptosis-related proteins. Our data revealed that PHMG-p reduced viability and induced morphological alterations in BEAS-2B cells. Exposure to PHMG-p induced excessive accumulation of mitochondrial ROS (mtROS) in BEAS-2B cells. PHMG-p activated caspase-dependent apoptosis as well as NLRP3/caspase-1/GSDMD-mediated- and caspase-3/GSDME-mediated pyroptosis through mitochondrial oxidative stress in BEAS-2B cells. Notably, PHMG-p reduced mitochondrial respiratory function and induced the translocation of Bax and cleaved GSDM into the mitochondria, leading to mitochondrial dysfunction. Our results enhanced our understanding of PHMG-p-induced lung toxicity by demonstrating that PHMG-p induces pyroptosis via mtROS-induced mitochondrial dysfunction in bronchial epithelial cells.


Subject(s)
Bronchi , Epithelial Cells , Guanidines , Mitochondria , Pyroptosis , Reactive Oxygen Species , Pyroptosis/drug effects , Humans , Reactive Oxygen Species/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Bronchi/drug effects , Bronchi/pathology , Bronchi/metabolism , Cell Line , Guanidines/toxicity , Cell Survival/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
20.
Respir Res ; 25(1): 212, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38762455

ABSTRACT

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.


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