Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 26.031
Filtrer
1.
Zhongguo Zhong Yao Za Zhi ; 49(14): 3868-3877, 2024 Jul.
Article de Chinois | MEDLINE | ID: mdl-39099360

RÉSUMÉ

The study investigated the effect of Compound Shougong Powder(CSGP) on the biological functions of triple-negative breast cancer(TNBC) cells and whether its mechanism of action was related to the epithelial-mesenchymal transition(EMT) signaling pathway. TNBC cells(MDA-MB-231 and BT-549) were treated with different concentrations of CSGP-containing serum. MTS assay was used to detect the effect of CSGP on the proliferation of TNBC cells. The EdU staining was used to detect the effect of CSGP on the proliferation of TNBC cells. Flow cytometry was used to examine the impact of CSGP on apoptosis of TNBC cells. Wound-healing and Transwell assays were used to evaluate the effects of different concentrations of CSGP on the migration and invasion capabilities of TNBC cells. RNA sequencing technology was utilized to elucidate its mechanism. Subsequently, qRT-PCR was performed to measure the mRNA expression levels of E-cadherin, N-cadherin, Slug, Snail, Vimentin, Twist, Zinc finger E-box-Binding homeobox 1(Zeb1), and Zinc finger E-box-Binding homeobox 2(Zeb2). Western blot was used to assess the protein expression levels of Slug, Vimentin, and E-cadherin. After intervention with CSGP, the proliferation of MDA-MB-231 and BT-549 cells significantly decreased, while the apoptosis rate markedly increased. The expression levels of the epithelial marker protein E-cadherin significantly increased, while the expression levels of the EMT-related transcription factors Slug and Vimentin showed a decrease. In conclusion, CSGP inhibits the EMT, thereby suppressing the malignant progression of TNBC.


Sujet(s)
Apoptose , Prolifération cellulaire , Médicaments issus de plantes chinoises , Transition épithélio-mésenchymateuse , Tumeurs du sein triple-négatives , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Humains , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Médicaments issus de plantes chinoises/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Apoptose/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Poudres/composition chimique , Cadhérines/génétique , Cadhérines/métabolisme
2.
Commun Biol ; 7(1): 940, 2024 Aug 03.
Article de Anglais | MEDLINE | ID: mdl-39097636

RÉSUMÉ

Endothelial cell physiology is governed by its unique microenvironment at the interface between blood and tissue. A major contributor to the endothelial biophysical environment is blood hydrostatic pressure, which in mechanical terms applies isotropic compressive stress on the cells. While other mechanical factors, such as shear stress and circumferential stretch, have been extensively studied, little is known about the role of hydrostatic pressure in the regulation of endothelial cell behavior. Here we show that hydrostatic pressure triggers partial and transient endothelial-to-mesenchymal transition in endothelial monolayers of different vascular beds. Values mimicking microvascular pressure environments promote proliferative and migratory behavior and impair barrier properties that are characteristic of a mesenchymal transition, resulting in increased sprouting angiogenesis in 3D organotypic model systems ex vivo and in vitro. Mechanistically, this response is linked to differential cadherin expression at the adherens junctions, and to an increased YAP expression, nuclear localization, and transcriptional activity. Inhibition of YAP transcriptional activity prevents pressure-induced sprouting angiogenesis. Together, this work establishes hydrostatic pressure as a key modulator of endothelial homeostasis and as a crucial component of the endothelial mechanical niche.


Sujet(s)
Jonctions adhérentes , Pression hydrostatique , Néovascularisation physiologique , Transduction du signal , Protéines de signalisation YAP , Animaux , Humains , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Jonctions adhérentes/métabolisme , Cadhérines/métabolisme , Cadhérines/génétique , Mouvement cellulaire , Cellules endothéliales/métabolisme , Cellules endothéliales/physiologie , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéines de signalisation YAP/métabolisme
3.
Pathol Oncol Res ; 30: 1611813, 2024.
Article de Anglais | MEDLINE | ID: mdl-39148954

RÉSUMÉ

Hereditary breast and ovarian cancer is a well-known genetic condition, inherited mainly in an autosomal dominant way, which elevates the risk of developing malignancies at a young age in heterozygous carriers. Advances in new generation sequencing have enabled medical professionals to determine whether a patient is harbouring mutations in moderate- or high penetrance susceptibility genes. We conducted a retrospective analysis among 275 patients who underwent genetic counselling and multigene panel testing for hereditary breast and ovarian cancer syndrome in our department. From these patients 74.5% (205/275) were affected by some type of malignancy, while the remaining 25.5% (70/275) had a positive family history of different cancers, suggesting a genetic predisposition. These tests confirmed a genetic variant in 29.8% and 28.6% of these patient groups respectively. The results also mirrored our general knowledge concerning the genetic background of hereditary breast and ovarian cancer, as variants in either one of the BRCA1 and BRCA2 genes proved to be the most common cause among our patients with 41.5%. Our test also detected a novel mutation in the CDH1 gene and three patients with double heterozygosity in two different susceptibility genes. This study demonstrates the relevance of genetic counselling and non-BRCA gene sequencing among cancer patients and patients who fulfil the criteria for genetic testing, while also providing important details about the genetic profile of Hungarian patients.


Sujet(s)
Protéine BRCA1 , Protéine BRCA2 , Prédisposition génétique à une maladie , Séquençage nucléotidique à haut débit , Mutation , Tumeurs de l'ovaire , Humains , Femelle , Études rétrospectives , Adulte , Adulte d'âge moyen , Protéine BRCA2/génétique , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Protéine BRCA1/génétique , Séquençage nucléotidique à haut débit/méthodes , Syndrome héréditaire de cancer du sein et de l'ovaire/génétique , Sujet âgé , Dépistage génétique/méthodes , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Conseil génétique , Jeune adulte , Marqueurs biologiques tumoraux/génétique , Antigènes CD , Cadhérines
4.
BMC Cardiovasc Disord ; 24(1): 431, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39148029

RÉSUMÉ

BACKGROUND: Hyperhomocysteinemia (HHcy) is an independent risk factor for atherosclerosis (AS). Endothelial mesenchymal transition (EndMT) refers to the process in which endothelial cells lose endothelial cell morphology and characteristic gene expression, and acquire phenotypic characteristics and gene expression related to mesenchymal cells. Numerous studies have confirmed that EndMT is involved in the formation of atherosclerosis. Catalpol is one of the active components of Rehmannia, which has antioxidant, anti-inflammatory, anti-tumor, neuroprotective and other biological activities. Studies have shown that catalpol can reduce atherosclerotic plaque induced by high sugar or fat. However, the effect of catalpol on HHCY-induced EndMT is unclear. METHODS AND RESULTS: In vitro HHcy-treated primary human umbilical vein endothelial cells (HUVECs) were used to construct a cell model, and the antioxidants N-acetylcysteine (NAC) and catalase alcohol were administered. In vivo C57BL/6N mice were given a diet fed with 4.4% high methionine chow to construct a HHcy mice model and were treated with catalpol. The results showed that hhcy could induce morphological transformation of endothelial cells into mesenchymal cells, increase intracellular ROS content, up-regulate α-SMA, N-cadherin, p-p65 protein expression, down-regulate VE-cadherin, CD31 protein expression, induce pathological changes of aortic root endothelium, and increase aortic endothelial ROS content. Catalpol reversed these hhcy induced outcomes. CONCLUSIONS: Catalpol inhibits HHcy-induced EndMT, and the underlying mechanism may be related to the ROS/NF-κB signaling pathway. Catalpol may be a potential drug for the treatment of HHcy-related AS.


Sujet(s)
Modèles animaux de maladie humaine , Cellules endothéliales de la veine ombilicale humaine , Hyperhomocystéinémie , Glucosides d'iridoïdes , Souris de lignée C57BL , Facteur de transcription NF-kappa B , Espèces réactives de l'oxygène , Transduction du signal , Animaux , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/anatomopathologie , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Glucosides d'iridoïdes/pharmacologie , Espèces réactives de l'oxygène/métabolisme , Hyperhomocystéinémie/traitement médicamenteux , Hyperhomocystéinémie/métabolisme , Hyperhomocystéinémie/complications , Cellules cultivées , Facteur de transcription NF-kappa B/métabolisme , Antioxydants/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Mâle , Cadhérines/métabolisme , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Athérosclérose/anatomopathologie , Athérosclérose/métabolisme , Athérosclérose/traitement médicamenteux , Athérosclérose/prévention et contrôle , Athérosclérose/étiologie , Facteur de transcription RelA/métabolisme , Antigènes CD/métabolisme ,
6.
Anal Chem ; 96(32): 13270-13277, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39093913

RÉSUMÉ

Epithelial-mesenchymal transition (EMT) is a complex process that plays a critical role in tumor progression. In this study, we present an EMT sensing panel for the classification of cancer cells at different EMT stages. This sensing panel consists of three types of fluorescent probes based on boronic acid-functionalized carbon-nitride nanosheet (BCN) derivatives. The selective response toward different EMT-associated biomarkers, namely, EpCAM, N-cadherin, and sialic acid (SA), was achieved by conjugating the corresponding antibodies to each BCN derivative, whereas the rare-earth-doping ensures simultaneous sensing of the three biomarkers with fluorescent emission of the three probes at different wavelengths. Sensitive sensing of the three biomarkers was achieved at the protein level with LODs reaching 1.35 ng mL-1 for EpCAM, 1.62 ng mL-1 for N-cadherin, and 1.54 ng mL-1 for SA. The selective response of these biomarkers on the cell surface also facilitated sensitive detection of MCF-7 cells and MDA-MB-231 cells with LODs of 2 cells/mL and 2 cells/mL, respectively. Based on the simultaneous sensing of the three biomarkers on cancer cells that underwent different extents of EMT, precise discrimination and classification of cells at various EMT stages were also achieved with an accuracy of 93.3%. This EMT sensing panel provided a versatile tool for monitoring the EMT evolution process and has the potential to be used for the evaluation of the EMT-targeting therapy and metastasis prediction.


Sujet(s)
Marqueurs biologiques tumoraux , Cadhérines , Transition épithélio-mésenchymateuse , Humains , Marqueurs biologiques tumoraux/analyse , Marqueurs biologiques tumoraux/métabolisme , Cadhérines/analyse , Cadhérines/métabolisme , Colorants fluorescents/composition chimique , Lignée cellulaire tumorale , Molécule d'adhérence des cellules épithéliales/métabolisme , Cellules MCF-7 , Acides boroniques/composition chimique , Acide N-acétyl-neuraminique/analyse , Acide N-acétyl-neuraminique/métabolisme
7.
Sci Rep ; 14(1): 18553, 2024 08 09.
Article de Anglais | MEDLINE | ID: mdl-39122790

RÉSUMÉ

Increased protein-bound uremic toxins (PBUTs) in patients with chronic kidney disease (CKD) are associated with cardiovascular diseases (CVDs); however, whether retention of PBUTs causes CVD remains unclear. Previous studies assessing the impacts of PBUTs on the vasculature have relied on 2D cell cultures lacking in vivo microenvironments. Here, we investigated the impact of various PBUTs (p-cresol (PC), indoxyl sulfate (IS), and p-cresyl sulfate (PCS)) on microvascular function using an organ-on-a-chip (OOC). Human umbilical vein endothelial cells were used to develop 3D vessels. Chronic exposure to PC resulted in significant vascular leakage compared with controls, whereas IS or PCS treatment did not alter the permeability of 3D vessels. Increased permeability induced by PC was correlated with derangement of cell adherens junction complex, vascular endothelial (VE)-cadherin and filamentous (F)-actin. Additionally, PC decreased endothelial viability in a concentration-dependent manner with a lower IC50 in 3D vessels than in 2D cultures. IS slightly decreased cell viability, while PCS did not affect viability. PC induced inflammatory responses by increasing monocyte adhesion to endothelial surfaces of 3D vessels and IL-6 production. In conclusion, this study leveraged an OOC to determine the diverse effects of PBUTs, demonstrating that PC accumulation is detrimental to ECs during kidney insufficiency.


Sujet(s)
Crésols , Cellules endothéliales de la veine ombilicale humaine , Inflammation , Humains , Crésols/métabolisme , Crésols/toxicité , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie , Indican/métabolisme , Indican/toxicité , Cadhérines/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Toxines urémiques/métabolisme , Perméabilité capillaire/effets des médicaments et des substances chimiques , Laboratoires sur puces , Sulfates organiques/métabolisme
8.
Zool Res ; 45(5): 1048-1060, 2024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39147719

RÉSUMÉ

Extracellular membrane proteins are crucial for mediating cell attachment, recognition, and signal transduction in the testicular microenvironment, particularly germline stem cells. Cadherin 18 (CDH18), a type II classical cadherin, is primarily expressed in the nervous and reproductive systems. Here, we investigated the expression of CDH18 in neonatal porcine prospermatogonia (ProSGs) and murine spermatogonial stem cells (SSCs). Disruption of CDH18 expression did not adversely affect cell morphology, proliferation, self-renewal, or differentiation in cultured porcine ProSGs, but enhanced cell adhesion and prolonged cell maintenance. Transcriptomic analysis indicated that the down-regulation of CDH18 in ProSGs significantly up-regulated genes and signaling pathways associated with cell adhesion. To further elucidate the function of CDH18 in germ cells, Cdh18 knockout mice were generated, which exhibited normal testicular morphology, histology, and spermatogenesis. Transcriptomic analysis showed increased expression of genes associated with adhesion, consistent with the observations in porcine ProSGs. The interaction of CDH18 with ß-catenin and JAK2 in both porcine ProSGs and murine SSCs suggested an inhibitory effect on the canonical Wnt and JAK-STAT signaling pathways during CDH18 deficiency. Collectively, these findings highlight the crucial role of CDH18 in regulating cell adhesion in porcine ProSGs and mouse SSCs. Understanding this regulatory mechanism provides significant insights into the testicular niche.


Sujet(s)
Cadhérines , Adhérence cellulaire , Animaux , Mâle , Suidae , Adhérence cellulaire/physiologie , Souris , Cadhérines/métabolisme , Cadhérines/génétique , Souris knockout , Spermatogonies/métabolisme , Spermatogonies/physiologie , Testicule/métabolisme , Testicule/physiologie , Cellules souches germinales adultes/métabolisme , Cellules souches germinales adultes/physiologie , Régulation de l'expression des gènes , Cellules souches/physiologie , Cellules souches/métabolisme
9.
Zhonghua Zhong Liu Za Zhi ; 46(8): 755-763, 2024 Aug 23.
Article de Chinois | MEDLINE | ID: mdl-39143798

RÉSUMÉ

Objective: To explore the gene mutation characteristics and the relationship between gene mutations and long-term prognosis in clinical stage ⅠA lung adenocarcinoma patients. Methods: A retrospective analysis was conducted on 63 clinical stage ⅠA lung adenocarcinoma patients who underwent surgical resection at the Cancer Hospital of the Chinese Academy of Medical Sciences from January 2007 to October 2012, with documented postoperative recurrence or metastasis, as well as those who had a follow-up duration of 10 years or more without recurrence or metastasis. Whole exome sequencing (WES) technology was used to analyze the gene mutation profiles in tumor tissues and univariate and multivariate Cox regression analysis were used to clarify the influencing factors for patient prognosis. Results: After long term follow-up, 13 out of the 63 patients (21%) experienced recurrence or metastasis. WES technology analysis revealed that the most common tumor related gene mutations occurred in epidermal growth factor receptor (EGFR), with a mutation rate of 65.1% (41/63), followed by tumor protein p53 (TP53), fatatypical cadherin 1 (FAT1), low density lipoprotein receptor-related protein 1B (LRP1B), mechanistic target of rapamycin (MTOR), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit gamma (PIK3CG), and SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily A, member 4 (SMARCA4), with mutation rates of 30.2% (19/63), 20.6% (13/63), 15.9% (10/63), 15.9% (10/63), 15.9% (10/63), and 15.9% (10/63), respectively. Multivariate Cox regression analysis showed that PIK3CG mutations (HR=21.52, 95% CI: 3.19-145.01),smoothened (SMO) mutations (HR=35.28, 95% CI: 3.12-398.39), catenin beta 1 (CTNNB1) mutations (HR=332.86, 95% CI: 15.76-7 029.05), colony stimulating factor 1 receptor (CSF1R) mutations (HR=8 109.60, 95% CI: 114.19-575 955.17), and v-Raf murine sarcoma viral oncogene homolog B (BRAF) mutations (HR=23.65, 95% CI: 1.86-300.43) were independent risk factors affecting the prognosis of clinical stage ⅠA lung adenocarcinoma patients. Conclusions: PIK3CG, SMO, CTNNB1, CSF1R, BRAF gene mutations are closely related to long-term recurrence or metastasis in clinical stage ⅠA lung adenocarcinoma. Patients with these gene mutations should be given closer clinical attention.


Sujet(s)
Adénocarcinome pulmonaire , Récepteurs ErbB , Tumeurs du poumon , Mutation , Récidive tumorale locale , Stadification tumorale , Protéine p53 suppresseur de tumeur , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/chirurgie , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Études rétrospectives , Pronostic , Récepteurs ErbB/génétique , Protéine p53 suppresseur de tumeur/génétique , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Cadhérines/génétique , Cadhérines/métabolisme , bêta-Caténine/génétique , bêta-Caténine/métabolisme , , Études de suivi , Mâle , Femelle , Adulte d'âge moyen , Protéines de liaison à l'ADN , Récepteurs aux lipoprotéines LDL , Facteurs de transcription
10.
Cell Mol Life Sci ; 81(1): 333, 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39112663

RÉSUMÉ

Synaptic dysfunction is an early pathogenic event leading to cognitive decline in Huntington's disease (HD). We previously reported that the active ADAM10 level is increased in the HD cortex and striatum, causing excessive proteolysis of the synaptic cell adhesion protein N-Cadherin. Conversely, ADAM10 inhibition is neuroprotective and prevents cognitive decline in HD mice. Although the breakdown of cortico-striatal connection has been historically linked to cognitive deterioration in HD, dendritic spine loss and long-term potentiation (LTP) defects identified in the HD hippocampus are also thought to contribute to the cognitive symptoms of the disease. The aim of this study is to investigate the contribution of ADAM10 to spine pathology and LTP defects of the HD hippocampus. We provide evidence that active ADAM10 is increased in the hippocampus of two mouse models of HD, leading to extensive proteolysis of N-Cadherin, which has a widely recognized role in spine morphology and synaptic plasticity. Importantly, the conditional heterozygous deletion of ADAM10 in the forebrain of HD mice resulted in the recovery of spine loss and ultrastructural synaptic defects in CA1 pyramidal neurons. Meanwhile, normalization of the active ADAM10 level increased the pool of synaptic BDNF protein and activated ERK neuroprotective signaling in the HD hippocampus. We also show that the ADAM10 inhibitor GI254023X restored LTP defects and increased the density of mushroom spines enriched with GluA1-AMPA receptors in HD hippocampal neurons. Notably, we report that administration of the TrkB antagonist ANA12 to HD hippocampal neurons reduced the beneficial effect of GI254023X, indicating that the BDNF receptor TrkB contributes to mediate the neuroprotective activity exerted by ADAM10 inhibition in HD. Collectively, these findings indicate that ADAM10 inhibition coupled with TrkB signaling represents an efficacious strategy to prevent hippocampal synaptic plasticity defects and cognitive dysfunction in HD.


Sujet(s)
Protéine ADAM10 , Amyloid precursor protein secretases , Hippocampe , Maladie de Huntington , Potentialisation à long terme , Protéines membranaires , Récepteur trkB , Transduction du signal , Animaux , Protéine ADAM10/métabolisme , Protéine ADAM10/génétique , Maladie de Huntington/métabolisme , Maladie de Huntington/anatomopathologie , Souris , Amyloid precursor protein secretases/métabolisme , Amyloid precursor protein secretases/antagonistes et inhibiteurs , Hippocampe/métabolisme , Hippocampe/anatomopathologie , Récepteur trkB/métabolisme , Récepteur trkB/antagonistes et inhibiteurs , Potentialisation à long terme/effets des médicaments et des substances chimiques , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Facteur neurotrophique dérivé du cerveau/métabolisme , Modèles animaux de maladie humaine , Cadhérines/métabolisme , Épines dendritiques/métabolisme , Épines dendritiques/anatomopathologie , Neuroprotection , Mâle , Souris de lignée C57BL , Plasticité neuronale , Protein-tyrosine kinases/métabolisme , Protein-tyrosine kinases/antagonistes et inhibiteurs , Protein-tyrosine kinases/génétique , Souris knockout
11.
Article de Russe | MEDLINE | ID: mdl-39113443

RÉSUMÉ

Mutations in the human PCDH19 gene lead to epileptic encephalopathy of early childhood. It is characterized by the early onset of serial seizures, cognitive impairment and behavioral disorders (including autistic personality traits). In most cases, difficulties arise in selecting therapy due to pharmacoresistance. The pathogenesis of the disease is complex. The data available to us at the moment from numerous studies present the pathogenesis of «PCDH19 syndrome¼ as multi-level, affecting both the epigenetic support of cell life, and development of stem cells and progenitor cells in the process of neuroontogenesis, and the influence on the neurotransmitter mechanisms of the brain, and disruption of the formation of neural networks with an inevitable increase in the excitability of the cerebral cortex as a whole, and local changes in the highly labile regulatory structures of the hippocampal region. And it is not surprising that all these changes entail not only (and perhaps not so much) epileptization, but a profound disruption of the regulation of brain activity, accompanied by autism spectrum disorders, more profound disorders in the form of schizophrenia or cyclothymia, and the formation of delayed psychomotor development. A «side branch¼ of these pathogenetic processes can also be considered the participation of PCDH19 dysfunctions in certain variants of oncogenesis. The need for polypharmacy (in most cases) confirms the diversity of mechanisms involved in the pathogenesis of the disease and makes the prospects for the development of effective and rational treatment regimens very vague. Cautious optimism is caused only by attempts at relatively specific treatment with ganaxolone.


Sujet(s)
Cadhérines , Épilepsie , Mutation , Protocadhérines , Humains , Cadhérines/génétique , Épilepsie/traitement médicamenteux , Épilepsie/génétique , Polypharmacie , Trouble du spectre autistique/génétique , Trouble du spectre autistique/traitement médicamenteux , Encéphale
12.
J Transl Med ; 22(1): 734, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39103891

RÉSUMÉ

BACKGROUND: Atrial fibrillation (AF) is associated with increased risk of stroke and mortality. It has been reported that the process of atrial fibrosis was regulated by ß-catenin in rats with AF. However, pathophysiological mechanisms of this process in human with AF remain unclear. This study aims to investigate the possible mechanisms of ß-catenin in participating in the atrial fibrosis using human right atrial appendage (hRAA) tissues . METHODS: We compared the difference of ß-catenin expression in hRAA tissues between the patients with AF and sinus rhythm (SR). The possible function of ß-catenin in the development of AF was also explored in mice and primary cells. RESULTS: Firstly, the space between the membrane of the gap junctions of cardiomyocytes was wider in the AF group. Secondly, the expression of the gap junction function related proteins, Connexin40 and Connexin43, was decreased, while the expression of ß-catenin and its binding partner E-cadherin was increased in hRAA and cardiomyocytes of the AF group. Thirdly, ß-catenin colocalized with E-cadherin on the plasma membrane of cardiomyocytes in the SR group, while they were dissociated and accumulated intracellularly in the AF group. Furthermore, the expression of glycogen synthase kinase 3ß (GSK-3ß) and Adenomatous Polyposis Coli (APC), which participated in the degradation of ß-catenin, was decreased in hRAA tissues and cardiomyocytes of the AF group. Finally, the development of atrial fibrosis and AF were proved to be prevented after inhibiting ß-catenin expression in the AF model mice. CONCLUSIONS: Based on human atrial pathological and molecular analyses, our findings provided evidence that ß-catenin was associated with atrial fibrosis and AF progression.


Sujet(s)
Fibrillation auriculaire , Fibrose , Atrium du coeur , Myocytes cardiaques , bêta-Caténine , Humains , Fibrillation auriculaire/anatomopathologie , Fibrillation auriculaire/métabolisme , bêta-Caténine/métabolisme , Animaux , Atrium du coeur/métabolisme , Atrium du coeur/anatomopathologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Mâle , Glycogen synthase kinase 3 beta/métabolisme , Cadhérines/métabolisme , Jonctions communicantes/métabolisme , Adulte d'âge moyen , Souris , Femelle , Connexine 43/métabolisme , Souris de lignée C57BL , Sujet âgé
13.
Mol Biol Rep ; 51(1): 837, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39042337

RÉSUMÉ

BACKGROUND: MiR-21-5p is a highly expressed microRNA that plays an important role in various cancer-promoting processes, including anchorage-independent growth, invasion, migration metastasis, and drug resistance in lung cancer. Studies indicate that miR-21-5p may contribute to these processes by promoting epithelial-mesenchymal transition (EMT). Ras homolog gene family member B (RhoB), a gene downregulated by miR-21-5p, has also been linked to EMT in lung cancer. However, the role of the miR-21-5p/RhoB axis in EMT regulation in lung adenocarcinoma remains unclear. In this study, we aimed to investigate the regulatory role of the miR-21-5p/RhoB axis in EMT and related in vitro functional characteristics such as migration, invasion, cisplatin resistance, and the formation of tumor spheroids. METHODS AND RESULTS: A549 cells were transfected with the miR-21-5p inhibitor, RhoB siRNA, and their corresponding negative controls. Wound healing, transwell invasion, Methyl thiazole tetrazolium (MTT), and sphere formation assays were also performed to evaluate the migration, invasion, cisplatin resistance, and anchorage-independent growth of A549 cells. RT-qPCR was used to determine the mRNA expression levels of EMT markers. MiR-21-5p knockdown inhibited migration, invasion, cisplatin resistance, and sphere formation while upregulating E-cadherin and downregulating Slug. Furthermore, RhoB silencing restored EMT and related in vitro functional characteristics in A549 cells. CONCLUSIONS: Knockdown of miR-21-5p inhibits EMT and related in vitro functional characteristics by upregulating RhoB, suggesting that miR-21-5p may promote EMT through downregulation of RhoB.


Sujet(s)
Adénocarcinome pulmonaire , Mouvement cellulaire , Transition épithélio-mésenchymateuse , Régulation de l'expression des gènes tumoraux , Tumeurs du poumon , microARN , Protéine G RhoB , Humains , Transition épithélio-mésenchymateuse/génétique , microARN/génétique , microARN/métabolisme , Protéine G RhoB/génétique , Protéine G RhoB/métabolisme , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Régulation de l'expression des gènes tumoraux/génétique , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Mouvement cellulaire/génétique , Cellules A549 , Résistance aux médicaments antinéoplasiques/génétique , Cisplatine/pharmacologie , Régulation positive/génétique , Prolifération cellulaire/génétique , Lignée cellulaire tumorale , Techniques de knock-down de gènes , Invasion tumorale/génétique , Cadhérines/génétique , Cadhérines/métabolisme
14.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 59(7): 653-662, 2024 Jul 09.
Article de Chinois | MEDLINE | ID: mdl-38949133

RÉSUMÉ

Objective: To investigate the regulatory effect and mechanism of interleukin-22 (IL-22) on the gingival epithelial barrier in the context of periodontal inflammation. Methods: IL-22 knockout (IL-22 KO) mice were constructed, and periodontitis mice models were established through oral gavage with polymicrobial inoculation. DNAs were extracted from the oral plaques of IL-22 KO periodontitis mice group (n=7) and their wild-type littermates periodontitis group (n=7) to establish a periodontitis-related oral microbiota database"PD-RiskMicroDB", determining the relationship between changes in oral microbiota and microbial function in two groups using 16S rRNA sequencing results. Gingival epithelial cells (GEC) were cultured by modified trypsinization method, and were stimulated with 100 µg/L IL-22, Porphyromonas gingivalis (Pg) (multiplicity of infection:100), separately or together for 3 and 12 hours. The experimental groups were as follows: control group (no stimulation), IL-22 group, Pg group and Pg+IL-22 group. The expression of barrier protein E-cadherin in each group at 3 h was detected by immunofluorescence, real-time fluorescence quantitative PCR (RT-qPCR) and Western blotting. Fluorescein isothiocyanate-dextran-mediated epithelial cell permeability experiment was conducted to clarify the changes in permeability of GEC in each group at 3 and 12 h. The mRNA expressions of E-cadherin in the gingival epithelium of wild-type littermates periodontitis group and IL-22 KO periodontitis group were detected by RT-qPCR. Fifteen C57BL/6 wild-type mice were randomly divided into control group (n=5), periodontitis group (n=5) and periodontitis+IL-22 treatment group (n=5). RT-qPCR and immunohistochemistry (IHC) staining were used to detect the expression level of E-cadherin in the gingival epithelium of each group. Results: 16S rRNA sequencing results showed that the composition of oral microbiota changed in IL-22 KO periodontitis group, of which the abundance of bacterial genera related to periodontal tissue invasion was significantly increased (linear discriminant analysis score: 2.22, P=0.009), compared with wild-type littermates periodontitis group. In vitro cell experiments showed that after Pg infection for 3 hours, the cell connections of GEC in Pg group were interrupted, and the fluorescence intensity of E-cadherin was reduced in Pg group compared with the control group. Meanwhile, the mRNA and protein expression levels of E-cadherin (mRNA: 0.69±0.12; protein: 0.60±0.12) were downregulated compared with the control group [mRNA: 1.00±0.00 (P=0.043); protein: 1.04±0.08 (P=0.003)], respectively. The fluorescence intensity of E-cadherin in the Pg+IL-22 group was enhanced compared with Pg group, and expression levels of E-cadherin mRNA (1.16±0.10) and protein (0.98±0.07) in Pg+IL-22 group showed a significant increase compared with Pg group [mRNA: 0.69±0.12 (P=0.005); protein: 0.60±0.12 (P=0.007)]. The result of epithelial permeability test showed that there was no statistical difference in epithelial permeability among control group, Pg group, IL-22 group and Pg+IL-22 group with treatment for 3 hours (F=0.20, P=0.893). While when the treatment time turned to be 12 hours, the epithelial barrier permeability showed a significant increase in Pg group (1.39±0.15) compared with control group (1.00±0.00, P=0.027), and a decrease in Pg+IL-22 group (1.02±0.18) compared with Pg group (1.39±0.15, P=0.034). In vivo, the mRNA expression of E-cadherin in the gingival epithelium of IL-22 KO periodontitis group decreased significantly (0.32±0.21) compared with wild-type littermates periodontitis group (1.01±0.01) (t=5.70, P=0.005). Moreover, RT-qPCR and IHC staining results showed that the mRNA expression level of E-cadherin (0.40±0.07) and absorbance value of E-cadherin positive expression (0.02±0.00) in gingival epithelial tissue of periodontitis group were both significantly down-regulated compared with control group [mRNA: 1.00±0.00 (P=0.005); absorbance value of E-cadherin positive expression: 0.04±0.01 (P=0.006)]. Meanwhile, the mRNA expression level of E-cadherin (1.06±0.24) and the absorbance value of E-cadherin positive expression (0.03±0.01) were both observed increase in periodontitis+IL-22 treatment group compared with periodontitis group (P=0.003, P=0.039). Conclusions: IL-22 may exert a protective effect on the gingival epithelial barrier in an inflammatory environment by regulating the invasiveness of oral microbiota and the expression of host barrier protein.


Sujet(s)
Cadhérines , Gencive , , Interleukines , Souris knockout , Microbiote , Parodontite , Porphyromonas gingivalis , Animaux , Interleukines/métabolisme , Cadhérines/métabolisme , Gencive/cytologie , Gencive/métabolisme , Gencive/microbiologie , Souris , Parodontite/microbiologie , Parodontite/métabolisme , Cellules épithéliales/métabolisme , ARN ribosomique 16S
15.
Anesth Analg ; 139(2): 385-396, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39008867

RÉSUMÉ

BACKGROUND: Currently, clinical indicators for evaluating endothelial permeability in sepsis are unavailable. Endothelium-derived extracellular vesicles (EDEVs) are emerging as biomarkers of endothelial injury. Platelet endothelial cell adhesion molecule (PECAM) and vascular endothelial (VE)-cadherin are constitutively expressed endothelial intercellular adhesion molecules that regulate intercellular adhesion and permeability. Herein, we investigated the possible association between EDEVs expressing intercellular adhesion molecules (PECAM+ or VE-cadherin+ EDEVs) and endothelial permeability and sepsis severity. METHODS: Human umbilical vein endothelial cells (HUVECs) were stimulated with tumor necrosis factor alpha (TNF-α) directly or after pretreatment with permeability-modifying reagents such as angiopoietin-1, prostacyclin, or vascular endothelial growth factor (VEGF) to alter TNF-α-induced endothelial hyperpermeability. Endothelial permeability was measured using the dextran assay or transendothelial electrical resistance. Additionally, a prospective cross-sectional observational study was conducted to analyze circulating EDEV levels in patients with sepsis. EDEVs were examined in HUVEC culture supernatants or patient plasma (nonsepsis, n = 30; sepsis, n = 30; septic shock, n = 42) using flow cytometry. The Wilcoxon rank-sum test was used for comparisons between 2 groups. Comparisons among 3 or more groups were performed using the Steel-Dwass test. Spearman's test was used for correlation analysis. Statistical significance was set at P < .05. RESULTS: TNF-α stimulation of HUVECs significantly increased EDEV release and endothelial permeability. Pretreatment with angiopoietin-1 or prostacyclin suppressed the TNF-α-induced increase in endothelial permeability and inhibited the release of PECAM+ and VE-cadherin+ EDEVs. In contrast, pretreatment with VEGF increased TNF-α-induced endothelial permeability and the release of PECAM+ and VE-cadherin+ EDEVs. However, pretreatment with permeability-modifying reagents did not affect the release of EDEVs expressing inflammatory stimulus-inducible endothelial adhesion molecules such as E-selectin, intracellular adhesion molecule-1, or vascular cell adhesion molecule-1. The number of PECAM+ EDEVs on admission in the septic-shock group (232 [124, 590]/µL) was significantly higher (P = .043) than that in the sepsis group (138 [77,267]/µL), with an average treatment effect of 98/µL (95% confidence interval [CI], 2-270/µL), and the number of VE-cadherin+ EDEVs in the septic-shock group (173 [76,339]/µL) was also significantly higher (P = .004) than that in the sepsis group (81 [42,159]/µL), with an average treatment effect (ATE) of 79/µL (95% CI, 19-171/µL); these EDEV levels remained elevated until day 5. CONCLUSIONS: EDEVs expressing intercellular adhesion molecules (PECAM+ or VE-cadherin+ EDEVs) may reflect increased endothelial permeability and could be valuable diagnostic and prognostic markers for sepsis.


Sujet(s)
Antigènes CD , Cadhérines , Perméabilité capillaire , Vésicules extracellulaires , Cellules endothéliales de la veine ombilicale humaine , Sepsie , Indice de gravité de la maladie , Humains , Vésicules extracellulaires/métabolisme , Sepsie/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Mâle , Études prospectives , Antigènes CD/métabolisme , Femelle , Adulte d'âge moyen , Cadhérines/métabolisme , Sujet âgé , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Études transversales , Cellules cultivées , Angiopoïétine-1/métabolisme , Marqueurs biologiques/métabolisme , Marqueurs biologiques/sang , Antigènes CD31/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Endothélium vasculaire/métabolisme , Prostacycline/métabolisme
16.
Int J Mol Sci ; 25(13)2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-39000339

RÉSUMÉ

Epithelial-mesenchymal transition (EMT) refers to the transformation of polar epithelial cells into motile mesenchymal cells under specific physiological or pathological conditions, thus promoting the metastasis of cancer cells. Epithelial cadherin (E-cadherin) is a protein that plays an important role in the acquisition of tumor cell motility and serves as a key EMT epithelial marker. In the present study, AW01178, a small-molecule compound with potential therapeutic efficacy, was identified via in-cell Western high-throughput screening technology using E-cadherin as the target. The compound induced the upregulation of E-cadherin at both mRNA and protein levels and inhibited the EMT of breast cancer cells in vitro as well as metastasis in vivo. Mechanistically, AW01178 is a novel benzacetamide histone deacetylase inhibitor (HDACi) mainly targeting class I histone deacetylases. AW01178 promoted the transcription and expression of E-cadherin through enhancing the acetylation level of histone H3 in the E-cadherin promoter region, thereby inhibiting the metastasis of breast cancer cells. The collective findings support the potential utility of the novel HDACi compound identified in this study, AW01178, as a therapeutic drug for breast cancer and highlight its value for the future development of HDACi structures as anticancer drugs.


Sujet(s)
Tumeurs du sein , Cadhérines , Transition épithélio-mésenchymateuse , Inhibiteurs de désacétylase d'histone , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Inhibiteurs de désacétylase d'histone/pharmacologie , Humains , Tumeurs du sein/anatomopathologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Femelle , Animaux , Cadhérines/métabolisme , Cadhérines/génétique , Lignée cellulaire tumorale , Métastase tumorale , Souris , Mouvement cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe , Souris nude , Histone/métabolisme
17.
Mol Genet Genomic Med ; 12(7): e2496, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39056403

RÉSUMÉ

BACKGROUND: Hereditary diffuse gastric cancer (HDGC) (OMIM# 137215) is an autosomal dominant cancer syndrome associated with CDH1 (OMIM# 192090) mutations. Prophylactic total gastrectomy (PTG) is the most recommended preventive treatment when a pathogenic mutation is found. However, the increasing use of genetic testing has led to the identification of incidental CDH1 mutations in individuals without a family history of gastric cancer. It remains unclear whether these patients should undergo prophylactic total gastrectomy. METHODS: Germline DNA, obtained from peripheral blood, was analysed by NGS. RESULTS: A 47-year-old woman was diagnosed with high-grade serous ovarian carcinoma, FIGO stage IIIC, with a Homologous Recombination Deficiency (HRD) GIS status of 78 (positive, cut-off: 43). She received chemotherapy and niraparib treatment. A multigene panel test revealed no pathogenic mutations in BRCA1 (OMIM# 113705)/BRCA2 (OMIM# 600185) genes, but a de novo deletion of exon 16 in CDH1 was found incidentally. She had no previous family history of gastric or breast cancer. The patient was enrolled in a surveillance program involving periodic endoscopy and was diagnosed with diffuse gastric cancer through biopsies of a pale area in the antrum after 1 year of close endoscopic follow-up. CONCLUSION: This case presents supportive evidence for the pathogenic classification of the loss of the last exon of CDH1.


Sujet(s)
Antigènes CD , Cadhérines , Tumeurs de l'ovaire , Tumeurs de l'estomac , Humains , Femelle , Adulte d'âge moyen , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Antigènes CD/génétique , Cadhérines/génétique , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Gastrectomie , Mutation germinale
18.
Cell Signal ; 121: 111295, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38996955

RÉSUMÉ

Calpain2 is a conventional member of the non-lysosomal calpain protease family that has been shown to affect the dynamics of focal and cell-cell adhesions by proteolyzing the components of adhesion complexes. Here, we inactivated calpain2 using CRISPR/Cas9 in epithelial MDCK cells. We show that depletion of calpain2 has multiple effects on cell morphology and function. Calpain2-depleted cells develop epithelial shape, however, they cover a smaller area, and cell clusters are more compact. Inactivation of calpain2 enhanced restoration of transepithelial electrical resistance after calcium switch, decreased cell migration, and delayed cell scattering induced by HGF/SF. In addition, calpain2 depletion prevented morphological changes induced by ERK2 overexpression. Interestingly, proteolysis of several calpain2 targets, including E-cadherin, ß-catenin, talin, FAK, and paxillin, was not discernibly affected by calpain2 depletion. Taken together, these data suggest that calpain2 regulates the stability of cell-cell and cell-substratum adhesions indirectly without affecting the proteolysis of these adhesion complexes.


Sujet(s)
Calpain , Adhérence cellulaire , Cellules épithéliales , Calpain/métabolisme , Animaux , Chiens , Cellules rénales canines Madin-Darby , Cellules épithéliales/métabolisme , Cellules épithéliales/cytologie , Mouvement cellulaire , Cadhérines/métabolisme , Protéolyse , Facteur de croissance des hépatocytes/métabolisme , bêta-Caténine/métabolisme , Calcium/métabolisme , Mitogen-Activated Protein Kinase 1/métabolisme , Systèmes CRISPR-Cas
19.
Exp Cell Res ; 441(1): 114155, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39002689

RÉSUMÉ

At least one-third of patients with epithelial ovarian cancer (OC) present ascites at diagnosis and almost all have ascites at recurrence especially because of the propensity of the OC cells to spread in the abdominal cavity leading to peritoneal metastasis. The influence of ascites on the development of pre-metastatic niches, and on the biological mechanisms leading to cancer cell colonization of the mesothelium, remains poorly understood. Here, we show that ascites weakens the mesothelium by affecting the morphology of mesothelial cells and by destabilizing their distribution in the cell cycle. Ascites also causes destabilization of the integrity of mesothelium by modifying the organization of cell junctions, but it does not affect the synthesis of N-cadherin and ZO-1 by mesothelial cells. Moreover, ascites induces disorganization of focal contacts and causes actin cytoskeletal reorganization potentially dependent on the activity of Rac1. Ascites allows the densification and reorganization of ECM proteins of the mesothelium, especially fibrinogen/fibrin, and indicates that it is a source of the fibrinogen and fibrin surrounding OC spheroids. The fibrin in ascites leads to the adhesion of OC spheroids to the mesothelium, and ascites promotes their disaggregation followed by the clearance of mesothelial cells. Both αV and α5ß1 integrins are involved. In conclusion ascites and its fibrinogen/fibrin composition affects the integrity of the mesothelium and promotes the integrin-dependent implantation of OC spheroids in the mesothelium.


Sujet(s)
Ascites , Fibrine , Fibrinogène , Intégrine alpha5bêta1 , Tumeurs de l'ovaire , Sphéroïdes de cellules , Microenvironnement tumoral , Humains , Femelle , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/métabolisme , Ascites/anatomopathologie , Ascites/métabolisme , Intégrine alpha5bêta1/métabolisme , Sphéroïdes de cellules/métabolisme , Sphéroïdes de cellules/anatomopathologie , Fibrinogène/métabolisme , Fibrine/métabolisme , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/métabolisme , Tumeurs du péritoine/anatomopathologie , Carcinome épithélial de l'ovaire/métabolisme , Carcinome épithélial de l'ovaire/anatomopathologie , Lignée cellulaire tumorale , Récepteur vitronectine/métabolisme , Protéine G rac1/métabolisme , Adhérence cellulaire , Péritoine/anatomopathologie , Péritoine/métabolisme , Épithélium/métabolisme , Épithélium/anatomopathologie , Cadhérines/métabolisme , Cellules cancéreuses en culture
20.
Proc Natl Acad Sci U S A ; 121(29): e2319829121, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38976736

RÉSUMÉ

In the developing human brain, only 53 stochastically expressed clustered protocadherin (cPcdh) isoforms enable neurites from individual neurons to recognize and self-avoid while simultaneously maintaining contact with neurites from other neurons. Cell assays have demonstrated that self-recognition occurs only when all cPcdh isoforms perfectly match across the cell boundary, with a single mismatch in the cPcdh expression profile interfering with recognition. It remains unclear, however, how a single mismatched isoform between neighboring cells is sufficient to block erroneous recognitions. Using systematic cell aggregation experiments, we show that abolishing cPcdh interactions on the same membrane (cis) results in a complete loss of specific combinatorial binding between cells (trans). Our computer simulations demonstrate that the organization of cPcdh in linear array oligomers, composed of cis and trans interactions, enhances self-recognition by increasing the concentration and stability of cPcdh trans complexes between the homotypic membranes. Importantly, we show that the presence of mismatched isoforms between cells drastically diminishes the concentration and stability of the trans complexes. Overall, we provide an explanation for the role of the cPcdh assembly arrangements in neuronal self/non-self-discrimination underlying neuronal self-avoidance.


Sujet(s)
Cadhérines , Neurones , Isoformes de protéines , Humains , Neurones/métabolisme , Cadhérines/métabolisme , Isoformes de protéines/métabolisme , Isoformes de protéines/génétique , Communication cellulaire , Simulation numérique , Neurites/métabolisme , Membrane cellulaire/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE