Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 10.678
Filtrer
1.
PLoS One ; 19(7): e0302451, 2024.
Article de Anglais | MEDLINE | ID: mdl-38968258

RÉSUMÉ

Even with advanced plasmid and viral vectors, attaining copy numbers of multiple genes among different transfected cells is challenging. We achieved one gene expression from a single-copy gene in one cell using a transgene competition system, a combination of the Kazusa cDNA clones and our dual recombinase-mediated cassette exchange system. All 48 nuclear receptors were simultaneously expressed in one dish at the same expression level in HEK293 using this system, and the cell proliferation rate was compared. Significant differences were observed between cells transfected with CMV- or EF1 promoter-driven expression of the 48 nuclear receptors after 8 weeks. The EF1-NR1I2 cell line, which exhibited the highest increase from 2 to 8 weeks, showed 1.13-fold higher proliferation than the EF1-DsRed line. On the other hand, the EF1-NR4A1 cell line, which showed the maximum decrease at 8 weeks, showed 0.88-fold lower proliferation than the EF1-DsRed line. The results were confirmed in both our transgene competition system and long-term growth experiments. Our transgene competition system offers a wide-range, simple, and accurate cell competition method.


Sujet(s)
Prolifération cellulaire , Transgènes , Humains , Cellules HEK293 , Prolifération cellulaire/génétique , Expression des gènes/génétique , Dosage génique , Récepteurs cytoplasmiques et nucléaires/génétique , Récepteurs cytoplasmiques et nucléaires/métabolisme , Transfection , Régions promotrices (génétique) , Vecteurs génétiques/génétique
2.
Mol Biol Rep ; 51(1): 712, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38824221

RÉSUMÉ

INTRODUCTION: Coronary artery disease (CAD) in young adults can have devastating consequences. The cardiac developmental gene MEIS1 plays important roles in vascular networks and heart development. This gene effects on the regeneration capacity of the heart. Considering role of MEIS1 in cardiac tissue development and the progression of myocardial infarction this study investigated the expression levels of the MEIS1, HIRA, and Myocardin genes in premature CAD patients compared to healthy subjects and evaluated the relationships between these genes and possible inflammatory factors. METHODS AND RESULTS: The study conducted a case-control design involving 35 CAD patients and 35 healthy individuals. Peripheral blood mononuclear cells (PBMCs) were collected, and gene expression analysis was performed using real-time PCR. Compared with control group, the number of PBMCs in the CAD group exhibited greater MEIS1 and HIRA gene expression, with fold changes of 2.45 and 3.6. The expression of MEIS1 exhibited a negative correlation with IL-10 (r= -0.312) expression and positive correlation with Interleukin (IL)-6 (r = 0.415) and tumor necrosis factor (TNF)-α (r = 0.534) gene expression. Moreover, there was an inverse correlation between the gene expression of HIRA and that of IL-10 (r= -0.326), and a positive correlation was revealed between the expression of this gene and that of the IL-6 (r = 0.453) and TNF-α (r = 0.572) genes. CONCLUSION: This research demonstrated a disparity in expression levels of MEIS1, HIRA, and Myocardin, between CAD and healthy subjects. The results showed that, MEIS1 and HIRA play significant roles in regulating the synthesis of proinflammatory cytokines, namely, TNF-α and IL-6.


Sujet(s)
Maladie des artères coronaires , Protéine du site-1 d'intégration des virus myéloïdes écotropiques , Protéines nucléaires , Transactivateurs , Adulte , Femelle , Humains , Mâle , Adulte d'âge moyen , Études cas-témoins , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Maladie des artères coronaires/génétique , Expression des gènes/génétique , Régulation de l'expression des gènes/génétique , Interleukine-10/génétique , Interleukine-6/génétique , Interleukine-6/métabolisme , Agranulocytes/métabolisme , Protéine du site-1 d'intégration des virus myéloïdes écotropiques/génétique , Protéine du site-1 d'intégration des virus myéloïdes écotropiques/métabolisme , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Transactivateurs/génétique , Transactivateurs/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Facteur de nécrose tumorale alpha/génétique , Facteur de nécrose tumorale alpha/métabolisme
3.
Nat Commun ; 15(1): 4890, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38849352

RÉSUMÉ

The human brain has been implicated in the pathogenesis of several complex diseases. Taking advantage of single-cell techniques, genome-wide association studies (GWAS) have taken it a step further and revealed brain cell-type-specific functions for disease loci. However, genetic causal associations inferred by Mendelian randomization (MR) studies usually include all instrumental variables from GWAS, which hampers the understanding of cell-specific causality. Here, we developed an analytical framework, Cell-Stratified MR (csMR), to investigate cell-stratified causality through colocalizing GWAS signals with single-cell eQTL from different brain cells. By applying to obesity-related traits, our results demonstrate the cell-type-specific effects of GWAS variants on gene expression, and indicate the benefits of csMR to identify cell-type-specific causal effect that is often hidden from bulk analyses. We also found csMR valuable to reveal distinct causal pathways between different obesity indicators. These findings suggest the value of our approach to prioritize target cells for extending genetic causation studies.


Sujet(s)
Encéphale , Étude d'association pangénomique , Analyse de randomisation mendélienne , Obésité , Polymorphisme de nucléotide simple , Locus de caractère quantitatif , Humains , Obésité/génétique , Obésité/métabolisme , Encéphale/métabolisme , Analyse sur cellule unique/méthodes , Prédisposition génétique à une maladie/génétique , Causalité , Régulation de l'expression des gènes , Expression des gènes/génétique
4.
Mol Ecol ; 33(13): e17419, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38808559

RÉSUMÉ

The role of phenotypic plasticity during colonization remains unclear due to the shifting importance of plasticity across timescales. In the early stages of colonization, plasticity can facilitate persistence in a novel environment; but over evolutionary time, processes such as genetic assimilation may reduce variation in plastic traits such that species with a longer evolutionary history in an environment can show lower levels of plasticity than recent invaders. Therefore, comparing species in the early stages of colonization to long-established species provides a powerful approach for uncovering the role of phenotypic plasticity during different stages of colonization. We compared gene expression between low-dissolved oxygen (DO) and high-DO populations of two cyprinid fish: Enteromius apleurogramma, a species that has undergone a recent range expansion, and E. neumayeri, a long-established native species in the same region. We sampled tissue either immediately after capture from the field or after a 2-week acclimation under high-DO conditions, allowing us to test for both evolved and plastic differences in low-DO vs high-DO populations of each species. We found that most genes showing candidate-evolved differences in gene expression did not overlap with those showing plastic differences in gene expression. However, in the genes that did overlap, there was counter-gradient variation such that plastic and evolved gene expression responses were in opposite directions in both species. Additionally, E. apleurogramma had higher levels of plasticity and evolved divergence in gene expression between field populations. We suggest that the higher level of plasticity and counter-gradient variation may have allowed rapid genetic adaptation in E. apleurogramma and facilitated colonization. This study shows how counter-gradient variation may impact the colonization of divergent oxygen environments.


Sujet(s)
Cyprinidae , Oxygène , Animaux , Oxygène/métabolisme , Cyprinidae/génétique , Phénotype , Expression des gènes/génétique , Adaptation physiologique/génétique , Génétique des populations
5.
Hum Cell ; 37(4): 1156-1169, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38814517

RÉSUMÉ

To explore the effects of ß-Sitosterol upon hepatocellular carcinoma cell proliferation, apoptosis, migration, invasion, and epithelial-mesenchymal transition (EMT), and to investigate the underlying mechanism using network pharmacology. Human hepatocellular carcinoma cell lines (Huh-7 and HCCLM3) were expose to gradient concentrations of ß-Sitosterol (5 µg/mL, 10 µg/mL, and 20 µg/mL). Cell viability and proliferation were assessed using MTT, CCK-8, colony formation, and EdU assays.Flow cytometry was employed to evaluate cell cycle and apoptosis. Scratch and Transwell assays were performed, respectively, to detect cell migration and invasion. The levels of apoptosis-associated proteins (BAX, BCL2, and cleaved caspase3) as well as EMT-associated proteins (E-cadherin, N-cadherin, Snail, and Vimentin) were detected in Huh-7 and HCCLM3 cell lines using Western blot analysis. The drug target gene for ß-Sitosterol was screened via PubChem and subsequently evaluated for expression in the GSE112790 dataset. In addition, the expression level of glycogen synthase kinase 3 beta (GSK3B) within the Cancer Genome Atlas-Liver Hepatocellular Carcinoma (TCGA-LIHC) database was analyzed, along with its correlation to the survival outcomes of patients with hepatocellular carcinoma. The diagnostic efficiency of GSK3B was assessed by analyzing the ROC curve. Subsequently, Huh-7 and HCCLM3 cell lines were transfected with the overexpression vector of GSK3B and then treated with ß-Sitosterol to further validate the association between GSK3B and ß-Sitosterol. GSK3B demonstrated a significantly elevated expression in patients with hepatocellular carcinoma, which could predict hepatocellular carcinoma patients' impaired prognosis based on GEO dataset and TCGA database. GSK3B inhibitor (CHIR-98014) notably inhibited cell proliferation and invasion, promoted cell apoptosis and cell cycle arrest at G0/G1 phase in hepatocellular carcinoma cells. ß-Sitosterol treatment further promoted the efffects of GSK3B inhibitor on hepatocellular carcinoma cells. GSK3B overexpression has been found to enhance the proliferative and invasive capabilities of hepatocellular carcinoma cells. Furthermore it has been observed that GSK3B overexpression, it has been obsear can partially reverse the inhibitory effect of ß-Sitosterol upon hepatocellular. ß-Sitosterol suppressed hepatocellular carcinoma cell proliferation and invasion, and enhanced apoptosis via inhibiting GSK3B expression.


Sujet(s)
Apoptose , Carcinome hépatocellulaire , Prolifération cellulaire , Transition épithélio-mésenchymateuse , Glycogen synthase kinase 3 beta , Tumeurs du foie , Sitostérol , Humains , Sitostérol/pharmacologie , Glycogen synthase kinase 3 beta/métabolisme , Glycogen synthase kinase 3 beta/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/génétique , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Lignée cellulaire tumorale , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/génétique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/génétique , Expression des gènes/génétique , Expression des gènes/effets des médicaments et des substances chimiques , Phénotype , Invasion tumorale/génétique , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/génétique , Pharmacologie des réseaux , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
6.
Hum Cell ; 37(4): 1141-1155, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38700744

RÉSUMÉ

Esophageal squamous cell carcinoma (ESCC) is a common malignant tumor in East Asia. Hypoxia, a hallmark of solid tumors, significantly alters redox homeostasis inside tumor microenvironment. This alteration drives tumor proliferation, invasion, and metastasis, leading to poor prognostic outcomes. However, the role of hypoxia-related genes in ESCC remains poorly understood. We employed RNA sequencing to identify differentially expressed genes in ESCC. Clinical data, transcriptome profiles, and a hypoxia-related gene set were extracted from open-source databases. A prognostic model was constructed using least absolute shrinkage and selection operator (LASSO) regression, which was then validated through Cox regression analysis. Within this prognostic model, we pinpointed and investigated a key hypoxia-related gene affecting prognosis. The gene's expression was validated using real-time PCR and immunohistochemistry in both esophageal carcinoma and normal tissues. Tumor proliferation was examined through in vitro and in vivo assays, including the Cell Counting Kit-8, EdU, colony formation, and subcutaneous tumor models. A robust four-gene prognostic model (VBP1, BGN, CDKN1A, and PPFIA1) was successfully constructed and validated. Among these, VBP1 emerged as a key gene, exhibiting high expression levels that correlated with poor prognosis in ESCC. Functional experiments confirmed that VBP1 significantly accelerated tumor proliferation both in vitro and in vivo. VBP1 is identified as a pivotal gene within the hypoxia-related prognostic signature, and it significantly promotes tumor proliferation in ESCC.


Sujet(s)
Prolifération cellulaire , Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Humains , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/anatomopathologie , Prolifération cellulaire/génétique , Carcinome épidermoïde de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/anatomopathologie , Carcinome épidermoïde de l'oesophage/métabolisme , Pronostic , Expression des gènes/génétique , Lignée cellulaire tumorale , Hypoxie/génétique , Régulation de l'expression des gènes tumoraux/génétique , Inhibiteur p21 de kinase cycline-dépendante/génétique , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Transcriptome/génétique , Animaux , Hypoxie tumorale/génétique
8.
Neurobiol Aging ; 140: 93-101, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38761538

RÉSUMÉ

Platelet activation of protease-activated receptor 4 (PAR4) and thrombin are at the top of a chain of events leading to fibrin deposition, microinfarcts, blood-brain barrier disruption, and inflammation. We evaluated mRNA expression of the PAR4 gene F2RL3 in human brain and global cognitive performance in participants with and without cognitive impairment or dementia. Data were acquired from the Religious Orders Study (ROS) and the Rush Memory and Aging Project (MAP). F2RL3 mRNA was elevated in AD cases and was associated with worse retrospective longitudinal cognitive performance. Moreover, F2RL3 expression interacted with clinical AD diagnosis on longitudinal cognition whereas this relationship was attenuated in individuals without cognitive impairment. Additionally, when adjusting for the effects of AD neuropathology, F2RL3 expression remained a significant predictor of cognitive decline. F2RL3 expression correlated positively with transcript levels of proinflammatory markers including TNFα, IL-1ß, NFκB, and fibrinogen α/ß/γ. Together, these results reveal that F2RL3 mRNA expression is associated with multiple AD-relevant outcomes and its encoded product, PAR4, may play a role in disease pathogenesis.


Sujet(s)
Maladie d'Alzheimer , Expression des gènes , ARN messager , Récepteurs à la thrombine , Humains , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Récepteurs à la thrombine/génétique , Récepteurs à la thrombine/métabolisme , Mâle , Femelle , Sujet âgé de 80 ans ou plus , ARN messager/métabolisme , Expression des gènes/génétique , Sujet âgé , Dysfonctionnement cognitif/génétique , Dysfonctionnement cognitif/étiologie , Dysfonctionnement cognitif/métabolisme , Encéphale/métabolisme , Cognition , Inflammation/génétique , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/génétique , Interleukine-1 bêta/génétique , Interleukine-1 bêta/métabolisme , Fibrinogène/génétique , Fibrinogène/métabolisme , Facteur de nécrose tumorale alpha/génétique , Facteur de nécrose tumorale alpha/métabolisme , Médiateurs de l'inflammation/métabolisme
9.
Hum Antibodies ; 32(2): 67-74, 2024.
Article de Anglais | MEDLINE | ID: mdl-38788064

RÉSUMÉ

BACKGROUND: Tumor necrosis factor-α (TNFα) is a crucial physiologic regulator of immune responses, and several disorders have been associated with its dysregulation. OBJECTIVE: This study aimed to understand TNFα gene expression in adult patients with liver and pancreas disorders and examine the impact of TNFα-238 genotypes on this population. METHODS: At the Ibn Al-Baladi Hospital in Baghdad, blood samples were collected from forty patients who were diagnosed with beta thalassemia together with pancreatic disease, forty patients who were diagnosed with thalassemia together with liver disorder, and forty patients who were diagnosed with thalassemia without pancreas or liver disorder. For the purpose of establishing a control group, forty samples were collected from persons who were of the same age and gender and seemed to be in good health. All of these individuals were deemed to be older than 18 years old. Through the utilization of real-time polymerase chain reaction (PCR), the level of TNF-α gene expression was investigated and assessed. The T-ARMS-PCR method was performed for detection and genotyping of TNFα-238 in thalassemia patients and healthy control samples. RESULTS: The result showed that TNF α gene expression assessment showed that group B (thalassemia patients with liver disorder) had higher folding than other groups while the lowest gene expression was in group D (as control group). Furthermore, the relationship between TNFα gene expressions folding with TNFα-238 genotypes in beta thalassemia major patients, discovered a considerable increase at GA genotype patients in TNFα gene expression level, followed by AA genotype compared to the GG genotype. Furthermore, the results of the current study showed an association between the presence of the mutant (A) allele whether heterozygous (GA) and homozygous (AA) with the TNF-α gene expression in thalassemia patients with liver and pancreatic disorders. CONCLUSION: Based on the results, it can be concluded that there is a relationship between the presence of the mutant (A) allele, whether heterozygous (GA) or homozygous (AA) of TNF-α 238, and TNF-α gene expression in liver and pancreatic diseases as well as in patients with thalassemia.


Sujet(s)
Génotype , Maladies du foie , Maladies du pancréas , Facteur de nécrose tumorale alpha , bêta-Thalassémie , Humains , bêta-Thalassémie/génétique , Facteur de nécrose tumorale alpha/génétique , Adulte , Mâle , Femelle , Iraq , Maladies du foie/génétique , Maladies du pancréas/génétique , Polymorphisme de nucléotide simple/génétique , Jeune adulte , Études cas-témoins , Fréquence d'allèle , Expression des gènes/génétique , Adolescent , Prédisposition génétique à une maladie , Allèles
10.
Hum Cell ; 37(4): 1024-1038, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38691334

RÉSUMÉ

Osteoporosis (OP) is a highly prevalent disorder characterized by low bone mass that severely reduces patient quality of life. Although numerous treatments for OP have been introduced in clinic, many have side effects and high costs. Therefore, there is still an unmet need for optimal solutions. Here, raw signal analysis was used to identify potential high-risk factors for OP, and the biological functions and possible mechanisms of action (MOAs) of these factors were explored via gene set enrichment analysis (GSEA). Subsequently, molecular biological experiments were performed to verify and analyze the discovered risk factors in vitro and in vivo. PMAIP1 was identified as a potential risk factor for OP and significantly suppressed autophagy in osteoblasts via the AMPK/mTOR pathway, thereby inhibiting the proliferation and differentiation of osteoblasts. Furthermore, we constructed an ovariectomy (OVX) model of OP in rats and simultaneously applied si-PMAIP1 for in vivo interference. si-PMAIP1 upregulated the expression of LC3B and p-AMPK and downregulated the expression of p-mTOR, and these effects were reversed by the autophagy inhibitor. Micro-CT revealed that, si-PMAIP1 significantly inhibited the development of osteoporosis in OVX model rats, and this therapeutic effect was attenuated by treatment with an autophagy inhibitor. This study explored the role and mechanism of PMAIP1 in OP and demonstrated that PMAIP1 may serve as a novel target for OP treatment.


Sujet(s)
AMP-Activated Protein Kinases , Autophagie , Modèles animaux de maladie humaine , Ostéoblastes , Ostéoporose , Transduction du signal , Sérine-thréonine kinases TOR , Autophagie/génétique , Ostéoblastes/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/physiologie , Ostéoporose/métabolisme , Ostéoporose/génétique , Ostéoporose/étiologie , Animaux , AMP-Activated Protein Kinases/métabolisme , AMP-Activated Protein Kinases/physiologie , Transduction du signal/génétique , Rats , Femelle , Humains , Ovariectomie , Cellules cultivées , Expression des gènes/génétique , Différenciation cellulaire/génétique , Rat Sprague-Dawley , Prolifération cellulaire/génétique
11.
Hum Cell ; 37(4): 1107-1119, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38691335

RÉSUMÉ

Chimeric antigen receptor T (CART) cell therapy has demonstrated promising potential in the treatment of hematologic malignancies. However, its application to solid tumors is limited due to the restrictive nature of the tumor microenvironment, resulting in functional failure and poor persistence of CART cells. Overexpression of Bcl-2 in human CART cells (hCART) has been found to significantly enhance their anti-apoptotic effects both in vitro and in vivo. Nevertheless, the evaluation of hCART cells in preclinical studies has predominantly relied on immunodeficient mice xenograft tumor models, making it challenging to assess the impact of hCART cells on normal tissues and the immune system. We established a murine CART (mCART) that overexpresses Bcl-2 and targets the epidermal growth factor receptor variant III (EGFRvIII), named EGFRvIII·mCART-Bcl2. It demonstrated superior proliferation, cytotoxicity, and anti-apoptotic capabilities in vitro. In an immunocompetent mouse model of abdominal metastasis of colorectal cancer, EGFRvIII·mCART-Bcl2 exhibited improved survival of CART in the abdomen, increased tumor clearance, and significantly prolonged overall mouse survival. In summary, our study provides evidence that the introduction of Bcl-2 into mCART cells can enhance their therapeutic efficacy against solid tumors while ensuring safety.


Sujet(s)
Modèles animaux de maladie humaine , Récepteurs ErbB , Protéines proto-oncogènes c-bcl-2 , Récepteurs chimériques pour l'antigène , Animaux , Protéines proto-oncogènes c-bcl-2/génétique , Protéines proto-oncogènes c-bcl-2/métabolisme , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs ErbB/génétique , Souris , Humains , Immunothérapie adoptive/méthodes , Expression des gènes/génétique , Apoptose/génétique , Prolifération cellulaire/génétique , Tumeurs colorectales/thérapie , Tumeurs colorectales/génétique , Tumeurs colorectales/immunologie , Tumeurs colorectales/anatomopathologie , Thérapie cellulaire et tissulaire/méthodes , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Lignée cellulaire tumorale
12.
Hum Cell ; 37(4): 1056-1069, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38744794

RÉSUMÉ

Epithelial mesenchymal transition (EMT) occurring in retinal pigment epithelial cells (RPE) is a crucial mechanism that contributes to the development of age-related macular degeneration (AMD), a pivotal factor leading to permanent vision impairment. Long non-coding RNAs (lncRNAs) have emerged as critical regulators orchestrating EMT in RPE cells. In this study, we explored the function of the lncRNA CYTOR (cytoskeleton regulator RNA) in EMT of RPE cells and its underlying mechanisms. Through weighted correlation network analysis, we identified CYTOR as an EMT-related lncRNA associated with AMD. Experimental validation revealed that CYTOR orchestrates TGF-ß1-induced EMT, as well as proliferation and migration of ARPE-19 cells. Further investigation demonstrated the involvement of CYTOR in regulating the WNT5A/NFAT1 pathway and NFAT1 intranuclear translocation in the ARPE-19 cell EMT model. Mechanistically, CHIP, EMSA and dual luciferase reporter assays confirmed NFAT1's direct binding to CYTOR's promoter, promoting transcription. Reciprocally, CYTOR overexpression promoted NFAT1 expression, while NFAT1 overexpression increased CYTOR transcription. These findings highlight a mutual promotion between CYTOR and NFAT1, forming a positive feedback loop that triggers the EMT phenotype in ARPE-19 cells. These discoveries provide valuable insights into the molecular mechanisms of EMT and its association with AMD, offering potential avenues for targeted therapies in EMT-related conditions, including AMD.


Sujet(s)
Transition épithélio-mésenchymateuse , Rétrocontrôle physiologique , Dégénérescence maculaire , Facteurs de transcription NFATC , ARN long non codant , Épithélium pigmentaire de la rétine , Transition épithélio-mésenchymateuse/génétique , Humains , Épithélium pigmentaire de la rétine/métabolisme , Épithélium pigmentaire de la rétine/cytologie , Facteurs de transcription NFATC/métabolisme , Facteurs de transcription NFATC/génétique , ARN long non codant/physiologie , ARN long non codant/génétique , ARN long non codant/métabolisme , Dégénérescence maculaire/génétique , Dégénérescence maculaire/métabolisme , Dégénérescence maculaire/anatomopathologie , Dégénérescence maculaire/étiologie , Expression des gènes/génétique , Prolifération cellulaire/génétique , Mouvement cellulaire/génétique , Facteur de croissance transformant bêta-1/métabolisme , Transduction du signal/génétique , Transduction du signal/physiologie , Cellules épithéliales/métabolisme , Lignée cellulaire , Cellules cultivées
13.
Hum Cell ; 37(4): 904-916, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38743204

RÉSUMÉ

Mesenchymal stem/stromal cells (MSCs), originating from the mesoderm, represent a multifunctional stem cell population capable of differentiating into diverse cell types and exhibiting a wide range of biological functions. Despite more than half a century of research, MSCs continue to be among the most extensively studied cell types in clinical research projects globally. However, their significant heterogeneity and phenotypic instability have significantly hindered their exploration and application. Single-cell sequencing technology emerges as a powerful tool to address these challenges, offering precise dissection of complex cellular samples. It uncovers the genetic structure and gene expression status of individual contained cells on a massive scale and reveals the heterogeneity among these cells. It links the molecular characteristics of MSCs with their clinical applications, contributing to the advancement of regenerative medicine. With the development and cost reduction of single-cell analysis techniques, sequencing technology is now widely applied in fundamental research and clinical trials. This study aimed to review the application of single-cell sequencing in MSC research and assess its prospects.


Sujet(s)
Cellules souches mésenchymateuses , Médecine régénérative , Analyse sur cellule unique , Analyse sur cellule unique/méthodes , Analyse sur cellule unique/tendances , Humains , Cellules souches mésenchymateuses/cytologie , Médecine régénérative/méthodes , Médecine régénérative/tendances , Différenciation cellulaire/génétique , Expression des gènes/génétique
14.
Hum Cell ; 37(4): 1008-1023, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38753278

RÉSUMÉ

Reproductive aging is associated with altered stress response and many other menopausal symptoms. Little is known about the adrenal expression of the anti-aging protein Klotho or how it is modulated by estrogen in ovariectomized stressed rats. Fifty-six Wistar female rats were assigned into seven equal groups. Sham-operated (Sham), sham stressed (Sham/STS), ovariectomized (OVR), ovariectomized stressed (OVR/STS), ovariectomized stressed rosiglitazone-treated (OVR/STS/R), ovariectomized stressed estrogen-treated (OVR/STS/E), and ovariectomized stressed estrogen/GW9662 co-treated (OVR/STS/E/GW) groups. All stressed rats were subjected daily to a one-hour restraint stress test for 19 days. At the end of the experiment, blood was collected for serum corticosterone (CORT) analysis. Adrenal tissues were obtained and prepared for polymerase chain reaction (PCR) assay, hematoxylin and eosin (H&E), immunohistochemistry-based identification of Klotho and PPAR-γ, and Oil Red O (ORO) staining. The rise in serum CORT was negligible in the OVR/STS group, in contrast to the Sham/STS group. The limited CORT response in the former group was restored by estrogen and rosiglitazone and blocked by estrogen/GW9226 co-administration. ORO-staining revealed a more evident reduction in the adrenal fat in the OVR/STS group, which was reversed by estrogen and counteracted by GW. Also, there was a comparable expression pattern of Klotho and PPAR-γ in the adrenals. The adrenal Klotho decreased in the OVR/STS group, but was reversed by estrogen treatment. GW9226/estrogen co-treatment interfered with the regulatory effect of estrogen on Klotho. The study suggested modulation of the adrenal Kotho expression by estrogen, in the ovariectomized rats subjected to a restraint stress test. This estrogen-provided adrenal protection might be mediated by PPAR-γ activation.


Sujet(s)
Cortex surrénal , Oestrogènes , Glucuronidase , Protéines Klotho , Ovariectomie , Récepteur PPAR gamma , Rat Wistar , Animaux , Femelle , Glucuronidase/métabolisme , Glucuronidase/génétique , Cortex surrénal/métabolisme , Cortex surrénal/effets des médicaments et des substances chimiques , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/génétique , Rats , Contention physique , Expression des gènes/effets des médicaments et des substances chimiques , Expression des gènes/génétique , Corticostérone/sang , Stress psychologique/métabolisme , Stress physiologique , Rosiglitazone/pharmacologie , Modèles animaux de maladie humaine , Vieillissement/métabolisme , Modèles animaux
15.
Hum Cell ; 37(4): 1070-1079, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38750405

RÉSUMÉ

Human ATP-binding cassette transporter C11 (ABCC11) is a membrane protein exhibiting ATP-dependent transport activity for a variety of lipophilic anions including endogenous substances and xenobiotics such as anti-cancer agents. Accumulating evidence indicates that ABCC11 wild type is responsible for the high-secretion phenotypes in human apocrine glands including wet type of earwax and the risk of axillary osmidrosis. Also, a less-functional variant of ABCC11 was reportedly associated with a risk for drug-induced toxicity in humans. Thus, functional change in ABCC11 may affect individual's constitution and drug toxicity, which led us to reason that functional validation of genetic variations in ABCC11 should be of importance. Therefore, in addition to p.G180R (a well-characterized non-functional variant of ABCC11), we studied cellular expression and function of 10 variants of ABCC11. In this study, ABCC11 function was evaluated as an ATP-dependent transport of radio labeled-dehydroepiandrosterone sulfate using ABCC11-expressing plasma membrane vesicles. Except for p.G180R, other 10 variants were maturated as an N-linked glycoprotein and expressed on the plasma membrane. We found that six variants impaired the net cellular function of ABCC11. Among them, p.R630W was most influential. Including this identification of a significantly-dysfunctional variant, our findings will extend our understanding of genetic variations and biochemical features of ABCC11 protein.


Sujet(s)
Transporteurs ABC , Variation génétique , Maladies des glandes sudoripares , Humains , Transporteurs ABC/génétique , Transporteurs ABC/métabolisme , Maladies des glandes sudoripares/génétique , Maladies des glandes sudoripares/étiologie , Facteurs de risque , Glandes apocrines/métabolisme , Membrane cellulaire/métabolisme , Expression des gènes/génétique , Transport biologique/génétique , Adénosine triphosphate/métabolisme
16.
Hum Cell ; 37(4): 1091-1106, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38782857

RÉSUMÉ

Severe corneal cryoinjury can cause permanent corneal swelling and bullous keratopathy, one of the main reason for loss of sight. Mouse amniotic fluid mesenchymal stem cells (mAF-MSCs) can repair corneal damage caused by freezing; however, whether the exosomes derived from mAF-MSCs have the same repair effect is unknown. In this study, the mAF-MSC-exosomes were transplanted into the eyeballs of corneal cryoinjured mice. Histopathological examination showed that the mAF-MSC-exosomes improved the corneal structure and status of corneal epithelial cells in corneal cryoinjured mice. RRBS-sequencing showed that compared with the control group, four genes (Rpl13-ps6, miR-33, Hymai, and Plagl1), underwent DNA hypermethylation modification after mAF-MSC-exosomes treatment. The result of FISH indicated that miR-33-3p hybridization signals were enhanced in corneal epithelial cells from mice treated with mAF-MSC-exosomes. Semi-quantitative PCR and western blotting indicated that mAF-MSC-exosomes contained high levels of DNMT1 mRNA and protein. Additionally, luciferase report assays indicated that miR-33-3p overexpression in NIH-3T3 mouse embryonic fibroblast cells inhibited the activity of luciferase carrying a sequence from the 3' untranslated region of Bcl6. Moreover, BCL6 mRNA and protein levels in corneal tissues from mice treated with mAF-MSC-exosomes were higher than those in the control group. Therefore, our results suggested that mAF-MSC-exosomes could repair corneal cryoinjury by releasing DNMT1, which induced hypermethylation of the miR-33 promoter in corneal epithelial cells. Consequent downregulated miR-33 transcription upregulated Bcl6 expression, ultimately achieving the repair of corneal cryoinjury in mice.


Sujet(s)
DNA (Cytosine-5-)-methyltransferase 1 , Méthylation de l'ADN , Épithélium antérieur de la cornée , Exosomes , Cellules souches mésenchymateuses , microARN , Animaux , microARN/génétique , microARN/métabolisme , Souris , Épithélium antérieur de la cornée/anatomopathologie , Épithélium antérieur de la cornée/métabolisme , Méthylation de l'ADN/génétique , Exosomes/génétique , Exosomes/métabolisme , Cellules souches mésenchymateuses/métabolisme , DNA (Cytosine-5-)-methyltransferase 1/génétique , DNA (Cytosine-5-)-methyltransferase 1/métabolisme , Régions promotrices (génétique)/génétique , Lésions de la cornée/génétique , Lésions de la cornée/étiologie , Lésions de la cornée/thérapie , Lésions de la cornée/métabolisme , Cellules épithéliales/métabolisme , Expression des gènes/génétique , Congélation , Cellules NIH 3T3
17.
Hum Cell ; 37(4): 917-930, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38806940

RÉSUMÉ

Aquaporins (AQPs) are small, integral proteins facilitating water transport across plasma cell membranes in response to osmotic gradients. This family has 13 unique members (AQP0-12), which can also transport glycerol, urea, gases, and other salute small molecules. AQPs play a crucial role in the regulation of different cellular processes, including metabolism, migration, immunity, barrier function, and angiogenesis. These proteins are found to aberrantly overexpress in various cancers, including colorectal cancer (CRC). Growing evidence has explored AQPs as a potential diagnostic biomarker and therapeutic target in different cancers. However, there is no comprehensive review compiling the available information on the crucial role of AQPs in the context of colorectal cancer. This review highlights the significance of AQPs as the biomarker and regulator of tumor cells metabolism. In addition, the proliferation, angiogenesis, and metastasis of tumor cells related to AQPs expression as well as function are discussed. Understanding the AQPs prominent role in chemotherapy resistance is of great importance clinically.


Sujet(s)
Aquaporines , Carcinogenèse , Tumeurs colorectales , Résistance aux médicaments antinéoplasiques , Métastase tumorale , Néovascularisation pathologique , Humains , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/génétique , Aquaporines/métabolisme , Aquaporines/physiologie , Carcinogenèse/métabolisme , Carcinogenèse/génétique , Résistance aux médicaments antinéoplasiques/génétique , Marqueurs biologiques tumoraux/métabolisme , Prolifération cellulaire , Expression des gènes/génétique , Thérapie moléculaire ciblée
18.
STAR Protoc ; 5(2): 103051, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38700978

RÉSUMÉ

Phospholipids are important biomolecules for the study of lipidomics, signal transduction, biodiesel, and synthetic biology; however, it is difficult to synthesize and analyze phospholipids in a defined in vitro condition. Here, we present a protocol for in vitro production and quantification of phospholipids. We describe steps for preparing a cell-free system consisting of fatty acid synthesis and a gene expression system that synthesizes acyltransferases on liposomes. The whole reaction can be completed within a day and the products are quantified by liquid chromatography-mass spectrometry. For complete details on the use and execution of this protocol, please refer to Eto et al.1.


Sujet(s)
Système acellulaire , Acides gras , Phospholipides , Phospholipides/métabolisme , Phospholipides/biosynthèse , Acides gras/métabolisme , Acides gras/biosynthèse , Système acellulaire/métabolisme , Expression des gènes/génétique , Liposomes/métabolisme , Liposomes/composition chimique , Chromatographie en phase liquide/méthodes , Acyltransferases/génétique , Acyltransferases/métabolisme , Spectrométrie de masse/méthodes
19.
Nat Neurosci ; 27(6): 1064-1074, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38769152

RÉSUMÉ

Ancestral differences in genomic variation affect the regulation of gene expression; however, most gene expression studies have been limited to European ancestry samples or adjusted to identify ancestry-independent associations. Here, we instead examined the impact of genetic ancestry on gene expression and DNA methylation in the postmortem brain tissue of admixed Black American neurotypical individuals to identify ancestry-dependent and ancestry-independent contributions. Ancestry-associated differentially expressed genes (DEGs), transcripts and gene networks, while notably not implicating neurons, are enriched for genes related to the immune response and vascular tissue and explain up to 26% of heritability for ischemic stroke, 27% of heritability for Parkinson disease and 30% of heritability for Alzheimer's disease. Ancestry-associated DEGs also show general enrichment for the heritability of diverse immune-related traits but depletion for psychiatric-related traits. We also compared Black and non-Hispanic white Americans, confirming most ancestry-associated DEGs. Our results delineate the extent to which genetic ancestry affects differences in gene expression in the human brain and the implications for brain illness risk.


Sujet(s)
, Encéphale , Méthylation de l'ADN , Humains , /génétique , Encéphale/métabolisme , Femelle , Mâle , /génétique , Autopsie , Expression des gènes/génétique , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/ethnologie , Sujet âgé , Adulte d'âge moyen
20.
Cell Mol Biol Lett ; 29(1): 66, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38724931

RÉSUMÉ

The development of compact CRISPR systems has facilitated delivery but has concurrently reduced gene editing efficiency, thereby limiting the further utilization of CRISPR systems. Enhancing the efficiency of CRISPR systems poses a challenging task and holds significant implications for the advancement of biotechnology. In our work, we report a synthetic dual-antibody system that can stably exist in the intracellular environment, specifically inhibiting the functions of NF-κB and ß-catenin. This not only elevates the transgenic expression of the CRISPR system by suppressing the innate immune response within cells to enhance the gene editing efficiency but also demonstrates a notable tumor inhibitory effect. Based on the specific output expression regulation of CRISPR-CasΦ, we constructed a CRISPR-based gene expression platform, which includes sensor modules for detecting intracellular ß-catenin and NF-κB, as well as an SDA module to enhance overall efficiency. In vitro experiments revealed that the CRISPR-based gene expression platform exhibited superior CDK5 expression inhibition efficiency and specific cytotoxicity towards tumor cells. In vitro experiments, we found that CRISPR-based gene expression platforms can selectively kill bladder cancer cells through T cell-mediated cytotoxicity. Our design holds significant assistant potential of transgene therapy and may offer the capability to treat other diseases requiring transgene therapy.


Sujet(s)
Systèmes CRISPR-Cas , Édition de gène , Tumeurs de la vessie urinaire , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/thérapie , Tumeurs de la vessie urinaire/métabolisme , Humains , Systèmes CRISPR-Cas/génétique , Lignée cellulaire tumorale , Édition de gène/méthodes , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/génétique , Expression des gènes/génétique , Régulation de l'expression des gènes tumoraux , Clustered regularly interspaced short palindromic repeats/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...