Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Am J Cancer Res ; 14(4): 1904-1913, 2024.
Article in English | MEDLINE | ID: mdl-38726286

ABSTRACT

Addressing the critical challenge of early ovarian cancer (OC) detection, our study focuses on identifying novel biomarkers by analyzing preoperative peripheral blood exosomes from high-grade serous ovarian cancer (HGSC) patients and healthy controls. Utilizing high-performance liquid chromatography-mass spectrometry-based quantitative proteomics, we isolated and analyzed peripheral blood exosomes to identify differentially expressed proteins (DEPs). This comprehensive analysis, supported by gene ontology enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) database assessments, revealed 28 proteins with decreased abundance and 33 with increased abundance in HGSC patients compared to controls. Notably, Zinc Finger Protein 587B (ZNF587B) exhibited a significant reduction in abundance, confirmed by decreased mRNA and protein levels in HGSC and normal ovarian tissues, consistent with omes exosomal protein expression levels. Immunohistochemical staining further confirmed reduced ZNF587B protein levels in HGSC tissues. The significant correlation between ZNF587B expression levels and tumor stage underscores its potential as a valuable biomarker for early liquid biopsy screening of OC. Our findings suggest ZNF587B plays a crucial role in early HGSC detection, highlighting the importance of further research to validate its clinical utility and improve ovarian cancer patient outcomes.

2.
Eur J Med Res ; 29(1): 4, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38173013

ABSTRACT

BACKGROUND: Female fertility declines with increased maternal age, and this decline is even more rapid after the age of 35 years. Follicular fluid (FF) is a crucial microenvironment that plays a significant role in the development of oocytes, permits intercellular communication, and provides the oocytes with nutrition. Exosomes have emerged as being important cell communication mediators that are linked to age-related physiological and pathological conditions. However, the metabolomic profiling of FF derived exosomes from advanced age females are still lacking. METHODS: The individuals who were involved in this study were separated into two different groups: young age with a normal ovarian reserve and advanced age. The samples were analysed by using gas chromatography-time of flight mass spectrometry (GC-TOFMS) analysis. The altered metabolites were analysed by using Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis to identify the functions and pathways that were involved. RESULTS: Our data showed that metabolites in exosomes from FF were different between women of young age and women of advanced age. The set of 17 FF exosomal metabolites (P ≤ 0.05) may be biomarkers to differentiate between the two groups. Most of these differentially expressed metabolites in FF were closely involved in the regulation of oocyte number and hormone levels. CONCLUSIONS: In this study, we identified differences in the metabolites of exosomes from FF between women of young age and women of advanced age. These different metabolites were tightly related to oocyte count and hormone levels. Importantly, these findings elucidate the metabolites of the FF exosomes and provide a better understanding of the nutritional profiles of the follicles with age.


Subject(s)
Exosomes , Follicular Fluid , Female , Humans , Adult , Follicular Fluid/chemistry , Follicular Fluid/metabolism , Ovarian Follicle/metabolism , Oocytes/metabolism , Hormones/analysis , Hormones/metabolism
3.
Front Genet ; 13: 892766, 2022.
Article in English | MEDLINE | ID: mdl-35832197

ABSTRACT

Normal heart development is vital for maintaining its function, and the development process is involved in complex interactions between different cell lineages. How mammalian hearts develop differently is still not fully understood. In this study, we identified several major types of cardiac cells, including cardiomyocytes (CMs), fibroblasts (FBs), endothelial cells (ECs), ECs/FBs, epicardial cells (EPs), and immune cells (macrophage/monocyte cluster, MACs/MONOs), based on single-cell transcriptome data from embryonic hearts of both human and mouse. Then, species-shared and species-specific marker genes were determined in the same cell type between the two species, and the genes with consistent and different expression patterns were also selected by constructing the developmental trajectories. Through a comparison of the development stage similarity of CMs, FBs, and ECs/FBs between humans and mice, it is revealed that CMs at e9.5 and e10.5 of mice are most similar to those of humans at 7 W and 9 W, respectively. Mouse FBs at e10.5, e13.5, and e14.5 are correspondingly more like the same human cells at 6, 7, and 9 W. Moreover, the e9.5-ECs/FBs of mice are most similar to that of humans at 10W. These results provide a resource for understudying cardiac cell types and the crucial markers able to trace developmental trajectories among the species, which is beneficial for finding suitable mouse models to detect human cardiac physiology and related diseases.

4.
Stem Cells ; 39(4): 443-457, 2021 04.
Article in English | MEDLINE | ID: mdl-33426760

ABSTRACT

ERG1, a potassium ion channel, is essential for cardiac action potential repolarization phase. However, the role of ERG1 for normal development of the heart is poorly understood. Using the rat embryonic stem cells (rESCs) model, we show that ERG1 is crucial in cardiomyocyte lineage commitment via interactions with Integrin ß1. In the mesoderm phase of rESCs, the interaction of ERG1 with Integrin ß1 can activate the AKT pathway by recruiting and phosphorylating PI3K p85 and focal adhesion kinase (FAK) to further phosphorylate AKT. Activation of AKT pathway promotes cardiomyocyte differentiation through two different mechanisms, (a) through phosphorylation of GSK3ß to upregulate the expression levels of ß-catenin and Gata4; (b) through promotion of nuclear translocation of nuclear factor-κB by phosphorylating IKKß to inhibit cell apoptosis, which occurs due to increased Bcl2 expression. Our study provides solid evidence for a novel role of ERG1 on differentiation of rESCs into cardiomyocytes.


Subject(s)
ERG1 Potassium Channel/genetics , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Integrin beta1/genetics , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-akt/genetics , Animals , Apoptosis/genetics , Cell Differentiation , Class Ia Phosphatidylinositol 3-Kinase/genetics , Class Ia Phosphatidylinositol 3-Kinase/metabolism , ERG1 Potassium Channel/metabolism , Embryo, Mammalian , Embryonic Stem Cells/cytology , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Integrin beta1/metabolism , Myocytes, Cardiac/cytology , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Signal Transduction , beta Catenin/genetics , beta Catenin/metabolism
5.
Mol Ther Nucleic Acids ; 21: 696-711, 2020 Sep 04.
Article in English | MEDLINE | ID: mdl-32769060

ABSTRACT

Cardiomyocyte differentiation derived from embryonic stem cells (ESCs) is a complex process involving molecular regulation of multiple levels. In this study, we first identify and compare differentially expressed gene (DEG) signatures of ESC-derived cardiomyocyte differentiation (ESCDCD) in humans and mice. Then, the multiscale embedded gene co-expression network analysis (MEGENA) of the human ESCDCD dataset is performed to identify 212 significantly co-expressed gene modules, which capture well the regulatory information of cardiomyocyte differentiation. Three modules respectively involved in the regulation of stem cell pluripotency, Wnt, and calcium pathways are enriched in the DEG signatures of the differentiation phase transition in the two species. Three human-specific cardiomyocyte differentiation phase transition modules are identified. Moreover, the potential regulation mechanisms of transcription factors during cardiomyocyte differentiation are also illustrated. Finally, several novel key drivers of ESCDCD are identified with the evidence of their expression during mouse embryonic cardiomyocyte differentiation. Using an integrative network analysis, the core molecular signatures and gene subnetworks (modules) underlying cardiomyocyte lineage commitment are identified in both humans and mice. Our findings provide a global picture of gene-gene co-regulation and identify key regulators during ESCDCD.

6.
Cell Signal ; 74: 109716, 2020 10.
Article in English | MEDLINE | ID: mdl-32707074

ABSTRACT

Cardiac dysfunction represents a main component of death induced by sepsis in critical care units. And microRNAs (miRNAs) have been reported as important modulators or biomarkers of sepsis. However, the molecular detail of miRNAs involved in septic cardiac dysfunction remains unclear. Here we showed that endotoxin (lipopolysaccharide, LPS) significantly down-regulated expression of miR-29b-3p in heart. Increased expression of miR-29b-3p by lentivirus improved cardiac function and attenuated damage of cardiac induced by LPS in mice. Furthermore, overexpression or knockdown of miR-29b-3p showed its crucial roles on regulation of apoptosis and production of pro-inflammatory cytokines in NRCMs through directly targeting FOXO3A. miR-29b-3p ameliorates inflammatory damage likely via reducing activation of MAPKs and nuclear-translocation of NF-κB to block LPS-activated NF-κB signaling. Notably, miR-29b is also down-regulated in septic patients' plasma compared with normal subjects, indicating a potential clinical relevance of miR-29b. Taken together, our findings demonstrate that upregulation of miR-29b-3p can attenuate myocardial injury induced by sepsis via regulating FOXO3A, which provide a potential therapy target for interference of septic cardiac dysfunction.


Subject(s)
Forkhead Box Protein O3/metabolism , Heart Diseases/metabolism , Inflammation/metabolism , Myocytes, Cardiac , Animals , Animals, Newborn , Apoptosis , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , MicroRNAs/physiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley
7.
Diabetes Res Clin Pract ; 151: 114-119, 2019 May.
Article in English | MEDLINE | ID: mdl-30935929

ABSTRACT

PURPOSE: The objective of this study was to estimate the combined effect of serum ferritin (SF) concentration and free thyroxine (fT4) levels on the risk of gestational diabetes mellitus (GDM). METHODS: Women presented for antenatal care at a tertiary hospital in Shanghai, China were included in this study from December 2012 to March 2014. Women were divided into six groups according to the SF and fT4 level. Multiple logistical regression model was used to estimate odds ratio (OR) among different groups. Relative excess risk of interaction (RERI), the attributable proportion (AP) of the interaction and the synergy index (SI) were applied to evaluate the additive interaction of SF concentration and fT4 level. RESULTS: A total of 6542 qualifying pregnant women were included in this study. We observed that a high SF concentration in early pregnancy was related to an increased risk of GDM (OR = 1.21, 95%CI: 1.02-1.43); while a low fT4 level was not (OR = 1.18, 95%CI: 0.89-1.58). There is no addictive interaction between SF and fT4 level on the presence of GDM. CONCLUSIONS: The study suggests that only high serum ferritin concentration is associated with an increased risk of GDM in early pregnancy. The level of fT4 in early pregnancy might have no effect on the association between high SF and risk of GDM.


Subject(s)
Diabetes, Gestational/blood , Diabetes, Gestational/diagnosis , Ferritins/blood , Thyroid Function Tests/methods , Thyroxine/blood , Adult , Diabetes, Gestational/pathology , Female , Humans , Pregnancy , Pregnancy Trimester, First/blood , Young Adult
8.
Curr Stem Cell Res Ther ; 9(1): 36-45, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24102581

ABSTRACT

Human amniotic fluid stem cells (hAFSCs) can be readily isolated from human amniotic fluid and display multi-differentiation potential and immunomodulatory properties. The mechanism of hAFSCs immunoregulation has not been defined. Here, we explore the immunomodulatory effects of hAFSCs derived from human amniotic fluid and evaluate the role of IL-10 and the tryptophan-catabolizing enzyme indoleamine 2,3-dioxygenase (IDO) in mediating the immunosuppressive actions of hAFSCs. Flow cytometry showed that hAFSCs were positive for the mesenchymal stem cell markers CD29, CD44, CD105, HLA-ABC, and more than 84% of the hAFSCs were positive for SSEA-4, which is a typical marker of embryonic stem cell (ESCs), and negative for HLA-DR. The RT-PCR and immunostaining results revealed that the multipotent stem cells expressed OCT-4, Nanog, CD44, SOX2 and SSEA-1. In vitro differentiation assays demonstrated that hAFSCs underwent osteogenic differentiation. We examined the immunomodulatory function of hAFSCs using a co-culture system with phorbol 12-myristate 13-acetate (PMA) stimulated peripheral blood mononuclear cells (PBMCs). PBMC proliferation was suppressed by the hAFSCs in a dose-dependent manner. The inhibitory effect was caused by increased IL-10 and IDO induction after co-culture. Neutralizing the IL-10 activity or blocking the function of IDO partially abolished the immunosuppressive action of the hAFSCs. In conclusion, these results suggest that the hAFSCs possess immunomodulatory properties, and IL-10 and IDO are involved in immunosuppression by hAFSCs.


Subject(s)
Embryonic Stem Cells/physiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interleukin-10/metabolism , Leukocytes, Mononuclear/physiology , Amniotic Fluid/cytology , Antigens, CD/metabolism , Cell Proliferation , Cells, Cultured , Coculture Techniques , Humans , Immunologic Factors/metabolism , Immunosuppression Therapy , Interferon-gamma/metabolism
9.
Oncol Rep ; 27(6): 1873-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22469973

ABSTRACT

High levels of gonadotropin are a risk factor for ovarian cancer development. Aberrant gonadotropin levels benefit tumor angiogenesis, but the detailed mechanism is not clear. Therefore, the aim of this study was to investigate the molecular mechanism of high levels of luteinizing hormone (LH) on the promotion of tumor angiogenesis and to outline a feasible therapeutic strategy. Western blotting and immunofluorescence staining were used to determine the effect of LH on VEGF and slit2 expression and examine the signaling pathway involved in regulating the expression of both molecules. Real-time PCR was used to investigate the effect of metformin on LH induction of VEGF and slit2 expression. It was found that 50 mIU/ml LH significantly upregulated VEGF and slit2 expression, and activated the PI3K/AKT-mTOR signaling pathway. However, metformin inhibited the mTOR signaling pathway and further blocked LH-induced VEGF and slit2 expression. In conclusion, high levels of LH promote angiogenesis in ovarian cancer via the PI3K/AKT-mTOR pathway. However, metformin could inhibit tumor angiogenesis by blocking the mTOR signaling pathway.


Subject(s)
Luteinizing Hormone/pharmacology , Metformin/pharmacology , Neovascularization, Pathologic , Ovarian Neoplasms/pathology , TOR Serine-Threonine Kinases/metabolism , Cell Line, Tumor , Female , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Luteinizing Hormone/metabolism , Nerve Tissue Proteins/biosynthesis , Ovarian Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis
10.
Sheng Li Xue Bao ; 61(2): 175-84, 2009 Apr 25.
Article in English | MEDLINE | ID: mdl-19377830

ABSTRACT

Apoptosis of abnormal oocytes is essential for defective oocyte elimination during prepubertal ovary development, and the ubiquitin system regulates the cell apoptosis via the degradation of specific proteins. Ubiquitin carboxyl-terminal hydrolase L1 (UCH-L1) is a component of the ubiquitin system, and the UCH-L1-dependent apoptosis is important for spermatogenesis. In the present study, the change in the number of follicles and the expression of UCH-L1 in oocytes were determined in prepubertal mouse ovaries by immunohistochemical techniques. A significant decrease in the follicular pool was found in prepubertal mouse ovaries during the period of day 21 to day 28 after birth, and accordingly, the UCH-L1 protein expression was increased, to some degree in association with Jun activation domain-binding protein 1 (Jab1) and cyclin-dependent kinase inhibitor p27(Kipl). The increased UCH-L1 protein, together with the corresponding changes of Jab1 was detected in morphologically abnormal oocytes of prepubertal ovaries. Through the immunofluorescent colocalization, UCH-L1 was shown concentrating in abnormal oocytes, and a parallel change in Jab1 was also seen. The affinity analysis confirmed the interaction between UCH-L1 and Jab1 in ovaries. These results suggest that UCH-L1 plays an important role, possibly in association with Jab1 and p27(Kipl), in selective elimination of abnormal oocytes during mouse prepubertal development.


Subject(s)
Apoptosis , Oocytes/cytology , Ovary/enzymology , Ubiquitin Thiolesterase/metabolism , Animals , Female , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...