Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Hum Cell ; 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39007956

ABSTRACT

Abnormal functions of trophoblast cells are associated with the pathogenesis of preeclampsia (PE). Nuclear receptor subfamily 2 group F member 1 (NR2F1) acts as a transcriptionally regulator in many diseases, but its role in PE remains unknown. Hypoxia/reoxygenation (H/R)-stimulated HTR-8/SVneo cells were used to mimic PE injury in vitro. NR2F1 overexpression alleviated trophoblast apoptosis, as evidenced by the decreased number of TUNEL-positive cells and the downregulation of caspase 3 and caspase 9 expression in cells. NR2F1 overexpression increased the invasion and migration ability of HTR-8/SVneo cells, accompanied by increased protein levels of matrix metalloproteinase (MMP)-2 and MMP-9. mRNA-seq was applied to explore the underlying mechanism of NR2F1, identifying growth differentiation factor 15 (GDF15) as the possible downstream effector. Dual-luciferase reporter, ChIP-qPCR, and DNA pull-down assays confirmed that NR2F1 bound to the promoter of GDF15 and transcriptionally inhibited its expression. GDF15 overexpression increased apoptosis and decreased the ability of invasion and migration in HTR-8/SVneo cells expressing NR2F1. MAPK pathway was involved in the regulation of PE. Administration of p38 inhibitor, ERK inhibitor, and JNK inhibitor reversed the effect of simultaneous overexpression NR2F1 and GDF15 on trophoblast apoptosis, invasion, and migration. Our findings demonstrated that NR2F1 overexpression inhibited trophoblast apoptosis and promoted trophoblast invasion and migration. NR2F1 might negatively regulate GDF15 expression by binding to its promoter region, which further inhibited MAPK signaling pathway in PE. Our study highlights that NR2F1 might sever as a potential target in PE.

2.
J Hypertens ; 42(7): 1154-1162, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38690926

ABSTRACT

BACKGROUND: : Circular RNAs (circRNAs) have been shown to be extensively involved in preeclampsia progression. At present, the role of circ_0007445 in preeclampsia progression is not clear. METHODS: A total of 30 preeclampsia patients and 30 normal pregnant women were recruited in our study. The function of trophoblast cells was explored to clarify the role and mechanism of circ_0007445 on the preeclampsia progression. The expression of circ_0007445, microRNA (miR)-4432 and high temperature requirement A1 (HTRA1) was analyzed by quantitative real-time PCR. The proliferation, migration and invasion of trophoblast cells were determined by cell counting kit 8 assay, EdU assay, colony formation assay, flow cytometry, and transwell assay. Protein expression was examined by western blot analysis. Dual-luciferase reporter assay, RNA immunoprecipitation (RIP) assay and RNA pull-down assay were used to assess RNA interaction relationships. RESULTS: Our data suggested that circ_0007445 had increased expression in preeclampsia patients. Knockdown of circ_0007445 enhanced trophoblast cell proliferation, migration and invasion. MiR-4432 was lowly expressed in preeclampsia patients, and it could be sponged by circ_0007445. MiR-4432 inhibitor overturned the promotion effects of circ_0007445 knockdown on trophoblast cell functions. HTRA1 was highly expressed in preeclampsia patients, and it could be targeted by miR-4432. HTRA1 overexpression could also reverse the proliferation, migration and invasion of trophoblast cells promoted by miR-4432 mimic. In addition, circ_0007445 positively regulated HTRA1 through targeting miR-4432. CONCLUSION: :Our results suggested that circ_0007445 facilitated the development of preeclampsia by suppressing trophoblast cell function through miR-4432/HTRA1 axis.


Subject(s)
Cell Movement , Cell Proliferation , High-Temperature Requirement A Serine Peptidase 1 , MicroRNAs , Pre-Eclampsia , RNA, Circular , Trophoblasts , Humans , Female , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Trophoblasts/metabolism , High-Temperature Requirement A Serine Peptidase 1/genetics , High-Temperature Requirement A Serine Peptidase 1/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Pregnancy , Adult
3.
Exp Cell Res ; 435(2): 113949, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38266865

ABSTRACT

HECW1 belongs to ubiquitin ligase (E3) HECT family, and is found to be involved in tumorigenesis and tumor progression. However, the function of HECW1 in cervical cancer (CC) remains unknown. Clinical analysis showed that HECW1 is significantly decreased in CC tumor tissues. Ectopic expression of HECW1 suppressed cell growth, promoting cell cycle arrest and apoptosis in CC cells, while downregulation of HECW1 reversed these trends, impeded proliferation and accelerated cell cycle progression of CC cells. Overexpressing of HECW1 reduced mitochondrial membrane potential and the protein expression of voltage-dependent anion channel 1 (VDAC1). In addition, upregulation of HECW1 inhibited nuclear ß-catenin accumulation, downregulated ß-catenin/TCF/LEF-mediated transcriptional activity and the expression of downstream gene c-Myc, whereas inhibition of HECW1 received opposite results. Further results confirmed HECW1 affects the protein expression of dishevelled-1 (DVL1), a potent activator of Wnt/ß-catenin, and inhibition of HECW1 inhibited the ubiquitination of DVL1, upregulating its expression. Inhibition of DVL1 restrained the promotion effect of HECW1 suppression on cell proliferation. In vivo experiments also verified that HECW1 suppression promoted the tumor formation of CC cells. Summary, we demonstrated that HECW1 inhibits CC cell proliferation and tumor formation by downregulating DVL1 induced Wnt/ß-catenin signaling pathway activation.


Subject(s)
Uterine Cervical Neoplasms , Wnt Signaling Pathway , Female , Humans , Wnt Signaling Pathway/genetics , Cell Line, Tumor , Uterine Cervical Neoplasms/pathology , beta Catenin/genetics , beta Catenin/metabolism , Ubiquitination , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Nerve Tissue Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
4.
Cell Cycle ; 22(20): 2211-2228, 2023 10.
Article in English | MEDLINE | ID: mdl-37974391

ABSTRACT

The development of chemotherapy resistance is a major obstacle for cervical cancer (CC) patients. Exosome-mediated transfer of circular RNAs (circRNAs) was found to have relevance to the CC. This study is designed to explore the role and mechanism of exosomal circRNA synaptotagmin 15 (circSYT15) on cisplatin (DDP) resistance in CC. Cell proliferation ability and apoptosis rate were detected by Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU), colony formation, and flow cytometry assays. CircSYT15, microRNA-503-5p (miR-503-5p), Remodeling spacing factor 1 (RSF1) levels were detected by real-time quantitative polymerase chain reaction (RT-qPCR). Exosomes were analyzed by a transmission electron microscope and nanoparticle tracking analysis. CD63, CD81, TSC101, Bcl-2, Bax, C-caspase 3, and RSF1 protein levels were examined by western blot assay. The binding between miR-503-5p and circSYT15 or RSF1 was predicted by circBank or Starbase and then verified by a dual-luciferase reporter and RNA Immunoprecipitation (RIP). The biological role of exosomal circSYT15 in DDP resistance of CC in vivo. CircSYT15 was upregulated in the DDP-resistant CC cells and exosomes isolated from DDP-resistant CC cells. CircSYT15 knockdown repressed the proliferation and drug resistance of CC and induced apoptosis in CC cells. Exosomes shuttled circSYT15 act as a sponge to affect RSF1 expression, thereby promoting proliferation and drug resistance and repressing apoptosis of sensitive CC cells. Exosomal circSYT15 boost DDP resistance of cervical cancer in vivo. Exosome-mediated transfer of circSYT15 enhanced DDP resistance in CC partly by targeting the miR-503-5p/RSF1 axis, providing a foundation for future clinical applications of CC drug resistance.


Subject(s)
Exosomes , MicroRNAs , Uterine Cervical Neoplasms , Humans , Female , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , RNA, Circular/genetics , Cisplatin/pharmacology , Cisplatin/therapeutic use , Exosomes/genetics , Cell Proliferation/genetics , MicroRNAs/genetics , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Nuclear Proteins , Trans-Activators
5.
Aging (Albany NY) ; 15(17): 8744-8769, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37671947

ABSTRACT

Several studies have reported the role of CLCN4 in tumor progression. However, its mechanism remains to be thoroughly studied. The objective of this study was to explore the potential pathogenic role of CLCN4 in endometrial carcinoma (UCEC) with a better understanding of the pathological mechanisms involved. The potential roles of CLCN4 in different tumors were explored based on The Cancer Genome Atlas (TCGA), the expression difference, mutation, survival, pathological stage, Immunity subtypes, Immune infiltration, tumor microenvironment (TME), tumor mutation burden (TMB), microsatellite instability (MSI), mismatch repair (MMR) related to CLCN4 were analyzed. Then, the expression, prognosis, mutation, and functional enrichment of CLCN4 in UCEC were analyzed. Immunohistochemical experiment was used to verify the expression of CLCN4 in endometrial cancer tissues and normal tissues. In vitro, we knocked down of CLCN4 in HEC-1-A cells and performed CCK8, WB, RT-PCR, wound-healing, transwell assays to further validation of the molecular function. Results revealed that high expression of CLCN4 was observed in 20 cancer types of TCGA. CLCN4 expression correlates with poor survival in MESO, BLCA, THCA, especially UCEC tumors. CLCN4 expression was significantly associated with CD4+ T-cell infiltration, especially CD4+ Th1-cell. Immunohistochemical experiment reveals that CLCN4 is high expressed in endometrial tumors, in vitro experiment reveals that knockdown of CLCN4 inhibits the cells proliferation, migration and invasion. Our study is the first to offer a comprehensive understanding of the oncogenic roles of CLCN4 on different tumors. CLCN4 may become a potential biomarker in UCEC.


Subject(s)
Endometrial Neoplasms , Female , Humans , Endometrial Neoplasms/genetics , Biomarkers , Biological Assay , CD4-Positive T-Lymphocytes , Cell Proliferation/genetics , Tumor Microenvironment , Chloride Channels/genetics
7.
Ann Clin Lab Sci ; 53(2): 278-292, 2023 Mar.
Article in English | MEDLINE | ID: mdl-37094850

ABSTRACT

OBJECTIVE: Cancer stem cells (CSCs) are responsible for cervical cancer progression and decreased radiosensitivity of tumor cells. The present work is meant to illuminate the impacts of exportin 1 (XPO1) on the aggressive behaviors and radiosensitivity of cervical cancer stemness cells and make a deeper inquiry into its regulatory mechanism despite that XPO1 has been supported to elicit significant activities on multiple malignancies. METHODS: XPO1 and Rad21 expression in HeLa (CD44+) cells was tested by RT-qPCR and western blot. Cell viability was estimated via CCK-8 assay. Cell stemness was examined via sphere formation assay and western blot. Following radiation treatment, cell proliferation was judged by CCK-8 assay, western blot as well as EdU staining whereas TUNEL assay, RT-qPCR and western blot analysis appraised cell apoptosis. Cell radiosensitivity was assessed through clonogenic survival assay. The levels of DNA damage markers were tested by western blot and related kits. STRING database and Co-IP assay respectively predicted and testified the binding of XPO1 with Rad21. RT-qPCR and western blot also examined the expression of XPO1 cargoes. RESULTS: The experimental data corroborated that XPO1 and Rad21 were overexpressed in cervical cancer tissues and cells. XPO1 inhibitor KPT-330 impeded the stemness while elevated the radiosensitivity of HeLa (CD44+) cells. XPO1 bond to Rad21 and positively modulated Rad21 expression. Moreover, Rad21 elevation reversed the impacts of KPT-330 on the behaviors of cervical cancer stemness cells. CONCLUSION: To sum up, XPO1 might impact the aggressive behavior and radioresistance of cervical cancer stemness cells through binding with Rad21.


Subject(s)
Uterine Cervical Neoplasms , Female , Humans , Uterine Cervical Neoplasms/metabolism , Karyopherins/genetics , Karyopherins/metabolism , Apoptosis , Cell Proliferation , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Cell Cycle Proteins , Hyaluronan Receptors/metabolism , Exportin 1 Protein
8.
Eur J Med Chem ; 249: 115122, 2023 Mar 05.
Article in English | MEDLINE | ID: mdl-36680987

ABSTRACT

Neurotoxic α-Syn fibers, the main components of Lewy bodies, play a key role in the development of PD characterized by a progressive loss of dopaminergic neurons. Here, we designed and synthesized the hybrids of polyphenolic/quinone acids. The candidate compounds showed high α-Syn aggregation inhibitory activities in vitro with IC50 down to 1.6 µM. The inhibition went through the aggregation process by stabilizing the conformation of α-Syn proteostasis and preventing ß-sheets aggregation, especially in the lag phase. Furthermore, the candidate drugs could disintegrate the preformed varisized aggregates into pony-size aggregates and functional monomers and continually inhibit the re-aggregation. The activities of anti-aggregation and aggregates depolymerization result in the reduction of inclusions in neuron cells. The candidate drugs also show high anti-oxidation and low cytotoxicity. They finally repair the damaged neurons in 6-OHDA-lesioned C57 mice and significantly improve PD-like symptoms of the PD model mice. The hybrids are promising molecules for PD prevention and therapy.© 2022 Elsevier Masson SAS. All rights reserved.


Subject(s)
Parkinson Disease , Mice , Animals , Horses , Parkinson Disease/drug therapy , alpha-Synuclein , Lewy Bodies , Neurons , Benzoquinones
9.
Funct Integr Genomics ; 21(2): 205-214, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33543402

ABSTRACT

The dysfunction of placenta development is correlated to the defects of pregnancy and fetal growth. The detailed molecular mechanism of placenta development is not identified in humans due to the lack of material in vivo. Trophoblast (TB) lineage derived from human embryonic stem cells (hESCs) induced by bone morphogenetic protein 4 (BMP4) has been applied as a model for studying TB lineage specification in vitro. With the development of single-cell sequencing technology, it became possible to detect the transcriptome of the post-implantation embryo at unprecedented precision. In this study, we reanalyzed single-cell RNA-seq of post-implantation embryos derived from two separate groups and identified different subtypes of trophoblast cells and their marker, respectively. At the same time, we focused on the gene expression patterns of trophoblast-specific transcription factors in different models. Our analysis sheds new light on the transcription regulation mechanism of trophoblast differentiation at the early stage of pregnancy establishment in human.


Subject(s)
Bone Morphogenetic Protein 4/genetics , Placenta/metabolism , Placentation/genetics , Transcription Factors/genetics , Blastocyst/metabolism , Blastocyst/pathology , Cell Differentiation/genetics , Cell Lineage/genetics , Embryonic Development/genetics , Female , Gene Expression Regulation, Developmental/genetics , Humans , Placenta/pathology , Pregnancy , RNA-Seq , Transcriptome/genetics , Trophoblasts/metabolism , Trophoblasts/pathology
10.
J Cell Mol Med ; 25(6): 3031-3040, 2021 03.
Article in English | MEDLINE | ID: mdl-33543559

ABSTRACT

As a common malignancy in females with a higher incidence rate, epithelial ovarian cancer (EOC) is a heterogeneous disease with complexity and diversity in histology and therapeutic response. Although great progress has been made in diagnosis and therapeutic strategies, novel therapeutic strategies are required to improve survival. Although the promoting effect of mucin 16 (MUC16) on tumour progression has been reported, the potential mechanisms remain unclear. In our study, we reported that overexpression of MUC16 was significantly related to cell proliferation and disease progression in EOC. Results from clinical specimen analysis and cell experiment support this conclusion. Patients with a high MUC16 expression usually had a worse prognosis that those with a low expression. Cell proliferation ability was significantly decreased in EOC cell lines when the knockdown of MUC16. Further study shows that the function of MUC16 in cell proliferation is based on the regulation of glucose transporter 1 (GLUT1) expression. MUC16 can control glucose uptake by regulating GLUT1 in EOC cells, thereby promoting glycogen synthesis, so that tumour cells produce more energy for proliferation. This conclusion is based on two findings. First, the significant correlation between MUC16 and GLUT1 was verified by clinical specimen and TCGA data analysis. Then, alteration of MUC16 expression levels can affect the expression of GLUT1 and glucose uptake was also verified. Finally, this conclusion is further verified in vivo by tumour-bearing mice model. To summarize, our results suggest that MUC16 promotes EOC proliferation and disease progression by regulating GLUT1 expression.


Subject(s)
CA-125 Antigen/genetics , Carcinoma, Ovarian Epithelial/genetics , Gene Expression Regulation, Neoplastic , Glucose Transporter Type 1/genetics , Membrane Proteins/genetics , Animals , CA-125 Antigen/metabolism , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Glucose Transporter Type 1/metabolism , Humans , Immunohistochemistry , Membrane Proteins/metabolism , Mice
11.
Cell Transplant ; 30: 963689720983786, 2021.
Article in English | MEDLINE | ID: mdl-33588586

ABSTRACT

Cervical cancer is one of the diseases that seriously endanger women's health. Circular RNA plays an important role in regulating the occurrence and development of cervical cancer. Here, we investigated the mechanisms of circ SMARCA5 in the development of cervical cancer. Quantitative reverse transcriptase polymerase chain reaction (RT-qPCR) results showed that the expression of SMARCA5 was downregulated in cervical cancer tissues and cell lines. Then we found that overexpression of SMARCA5 inhibited proliferation and invasion, but promoted apoptosis in cervical cancer cells. These were detected by Cell Counting Kit-8, Transwell, and Annexin V-fluorescein isothiocyanate/propidium iodide detection kit, respectively, and the expression of the apoptosis-related proteins was determined by western blotting. Then we predicted that SMARCA5 combined with Staphylococcal nuclease domain-containing 1 (SND1) by starBase, and verified by RNA pull-down assay. To further reveal the molecular mechanisms of SMARCA5 in the progression of cervical cancer, the interaction protein of SND1 was predicted by STRING, and the interaction was verified by co-immunoprecipitation assay. Then, the effects of SND1 or YWHAB on the development of cervical cancer were detected by the gain and loss function test, and we found that knockdown of SND1 or YWHAB reversed the effects of SMARCA5 short interfering RNA on proliferation, invasion, and apoptosis of cervical cancer cells. Overexpression of SMARCA5 inhibited cervical cancer metastasis in vivo. Our results showed that overexpression of circ SMARCA5 inhibits the binding of SND1 to YWHAB, and inhibits the proliferation and invasion, but promotes apoptosis in cervical cancer cells, thus inhibiting the metastasis of cervical cancer.


Subject(s)
14-3-3 Proteins/metabolism , Endonucleases/metabolism , RNA, Circular/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , 14-3-3 Proteins/genetics , Animals , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Endonucleases/genetics , Female , Humans , Mice , Mice, Nude , Neoplasm Metastasis , RNA, Circular/genetics , Transfection , Uterine Cervical Neoplasms/pathology , Xenograft Model Antitumor Assays
12.
BMC Cancer ; 21(1): 163, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33588776

ABSTRACT

BACKGROUND: Ovarian cancer (OC) is a life-threatening gynecological malignancy where dysregulation of microRNAs (miRNAs) is frequently implicated. This study focuses on the function of miR-545 on OC development and the molecules involved. METHODS: miR-545 expression in OC tissues and cell lines was determined, and its link to the survival of patients was analyzed. Altered expression of miR-545 was induced to determine its role in proliferation, apoptosis, migration and invasion of OC cells and the angiogenesis ability of human umbilical vein endothelial cells (HUVECs). The targeting mRNAs of miR-545 were predicted and validated through luciferase assays. Gain-of-function studies of KDM4B and PLK1 were performed to explore their involvements in OC development. In vivo experiments were conducted by inducing xenograft tumors in nude mice. RESULTS: Poor expression of miR-545 was found in OC tissues and cells compared to the normal ones and it indicated unfavorable prognosis in patients. Overexpression of miR-545 suppressed growth, migration, invasion and angiogenesis of OC cells as well as the angiogenesis ability of HUVECs. miR-545 was found to target mRNAs of KDM4B and PLK1, while KDM4B promoted the transcription of the PLK1 promoter through demethylation of H3K9me3. Either overexpression of KDM4B or PLK1 partially blocked the inhibitory effects of miR-545 mimic on OC cell growth, especially the former one. The in vitro results were reproduced in vivo. CONCLUSION: This study evidenced that miR-545 suppresses progression of OC through mediating PLK1 expression by a direct binding and an indirect regulation involving KDM4B-mediated demethylation.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Cycle Proteins/metabolism , Demethylation , Gene Expression Regulation, Neoplastic , Jumonji Domain-Containing Histone Demethylases/metabolism , MicroRNAs/genetics , Ovarian Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Cycle Proteins/genetics , Cell Proliferation , Disease Progression , Female , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Prognosis , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
13.
Reprod Toxicol ; 99: 39-47, 2021 01.
Article in English | MEDLINE | ID: mdl-33249234

ABSTRACT

The use of models of stem cell differentiation to trophoblastic cells provides an effective perspective for understanding the early molecular events in the establishment and maintenance of human pregnancy. In combination with the newly developed deep learning technology, the automated identification of this process can greatly accelerate the contribution to relevant knowledge. Based on the transfer learning technique, we used a convolutional neural network to distinguish the microscopic images of Embryonic stem cells (ESCs) from differentiated trophoblast -like cells (TBL). To tackle the problem of insufficient training data, the strategies of data augmentation were used. The results showed that the convolutional neural network could successfully recognize trophoblast cells and stem cells automatically, but could not distinguish TBL from the immortalized trophoblast cell lines in vitro (JEG-3 and HTR8-SVneo). We compare the recognition effect of the commonly used convolutional neural network, including DenseNet, VGG16, VGG19, InceptionV3, and Xception. This study extends the deep learning technique to trophoblast cell phenotype classification and paves the way for automatic bright-field microscopic image analysis of trophoblast cells in the future.


Subject(s)
Bone Morphogenetic Protein 4 , Human Embryonic Stem Cells/cytology , Neural Networks, Computer , Trophoblasts/classification , Cell Differentiation , Cells, Cultured , Gene Expression Regulation , Humans , Microscopy, Phase-Contrast , Phenotype
14.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 37(10): 1139-1142, 2020 Oct 10.
Article in Chinese | MEDLINE | ID: mdl-32924120

ABSTRACT

OBJECTIVE: To explore the genetic basis of a fetus with enlargement and enhanced echo of the kidneys. METHODS: The imaging data of the fetus were collected, in addition with 20 mL amniotic fluid sample and 2 mL peripheral blood samples of both parents. Amniotic DNA was extracted for library construction and whole exome sequencing, and Sanger sequencing was carried out to verify candidate variant associated with the fetal phenotype. RESULTS: Prenatal ultrasound showed that the fetus had enlargement and enhanced echo of the kidneys, in addition with many small renal cysts. Whole exome sequencing showed that the fetus carried pathogenic compound heterozygous variants of the ETFDH gene, namely c.3G>C and c.1436dupA. Sanger sequencing of the family suggested that the variants were inherited from its mother and father, respectively. CONCLUSION: By combining its clinical manifestations and results of whole exome sequencing, the fetus was diagnosed as glutaric acidemia type ⅡC due to the compound heterozygous variants of the ETFDH gene. Above results have provided a basis for prenatal diagnosis and genetic counseling. Fetal exome sequencing has provided an important tool for prenatal diagnosis.


Subject(s)
Exome Sequencing , Fetus , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/diagnosis , Prenatal Diagnosis , DNA , Electron-Transferring Flavoproteins/genetics , Female , Humans , Iron-Sulfur Proteins/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Phenotype , Pregnancy
15.
Aging (Albany NY) ; 12(10): 9041-9065, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32409627

ABSTRACT

Sigma non-opioid intracellular receptor 1 (sigma-1 receptor), a non-opioid transmembrane protein, is located on cellular mitochondrial membranes and endoplasmic reticulum. Current research has demonstrated that sigma-1 receptor is related to human degenerative diseases. This study is focused on the effects of sigma-1 receptor on the pathophysiological process of diminished ovarian reserve (DOR) and granulosa cells (GCs) apoptosis. Sigma-1 receptor concentration in follicular fluid (FF) and serum were negatively correlated with basal follicle-stimulating hormone (FSH) and positively correlated with anti-mullerian hormone (AMH), antral follicle count (AFC). Sigma-1 receptor reduction in GCs was accompanied by endoplasmic reticulum stress (ERS)-mediated apoptosis in women with DOR. Plasmid transfection was used to establish SIGMAR1-overexpressed and SIGMAR1-knockdown human granulosa-like tumor (KGN) cell and thapsigargin (TG) was used to induce ERS KGN cells. We found that KGN cells treated with endogenous sigma-1 receptor ligand dehydroepiandrosterone (DHEA) and sigma-1 receptor agonist PRE-084 showed similar biological effects to SIGMAR1-overexpressed KGN cells and opposite effects to SIGMAR1-knockdown KGN cells. DHEA may improve DOR patients' pregnancy outcomes by upregulating sigma-1 receptor and downregulating ERS-mediated apoptotic genes in GCs. Thus, sigma-1 receptor may be a potential ovarian reserve biomarker, and ligand-mediated sigma-1 receptor activation could be a future approach for DOR therapy.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress/physiology , Granulosa Cells/physiology , Ovarian Reserve/physiology , Receptors, sigma , Adult , Apoptosis/genetics , Apoptosis/physiology , Female , Humans , Ovary/cytology , Ovary/metabolism , Ovary/pathology , Receptors, sigma/genetics , Receptors, sigma/metabolism , Sigma-1 Receptor
16.
Cancer Cell Int ; 19: 58, 2019.
Article in English | MEDLINE | ID: mdl-30923460

ABSTRACT

BACKGROUND: Tumor necrosis factor-α (TNF-α) immunotherapy controls the progression of human cervical cancer. Here, we explored the detailed molecular mechanisms played by melatonin in human cervical cancer (HeLa cells) death in the presence of TNF-α injury, with a particular attention to the mitochondrial homeostasis. METHODS: HeLa cells were incubated with TNFα and then cell death was determined via MTT assay, TUNEL staining, caspase ELISA assay and western blotting. Mitochondrial function was detected via analyzing mitochondrial membrane potential using JC-1 staining, mitochondrial oxidative stress using flow cytometry and mitochondrial apoptosis using western blotting. RESULTS: Our data exhibited that treatment with HeLa cells using melatonin in the presence of TNF-α further triggered cancer cell cellular death. Molecular investigation demonstrated that melatonin enhanced the caspase-9 mitochondrion death, repressed mitochondrial potential, increased ROS production, augmented mPTP opening rate and elevated cyt-c expression in the nucleus. Moreover, melatonin application further suppressed mitochondrial ATP generation via reducing the expression of mitochondrial respiratory complex. Mechanistically, melatonin augmented the response of HeLa cells to TNF-α-mediated cancer death via repressing mitophagy. TNF-α treatment activated mitophagy via elevating Parkin expression and excessive mitophagy blocked mitochondrial apoptosis, ultimately alleviating the lethal action of TNF-α on HeLa cell. However, melatonin supplementation could prevent TNF-α-mediated mitophagy activation via inhibiting Parkin in a CaMKII-dependent manner. Interestingly, reactivation of CaMKII abolished the melatonin-mediated mitophagy arrest and HeLa cell death. CONCLUSIONS: Overall, our data highlight that melatonin enhances TNF-α-induced human cervical cancer HeLa cells mitochondrial apoptosis via inactivating the CaMKII/Parkin/mitophagy axis.

17.
Oncol Lett ; 17(1): 539-547, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30655799

ABSTRACT

In the present study, differentially expressed microRNAs (miRNAs) in peritoneal exosomes that were isolated from 10 patients with epithelial ovarian cancer (EOC) with metastasis in the abdominal cavity and 10 participants without cancer (NC) were identified. These differentially expressed miRNAs that were revealed by next-generation sequencing were categorized by Gene Ontology enrichment and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis of their target genes. Notably, two miRNAs that were associated with EOC-miR-149-3p and miR-222-5p-were identified. There were significant differences in expression of miR-149-3p and miR-222-5p between EOC and NC samples, and the effect of the expression level of the two miRNAs on the patient survival was identified using publicly available data from The Cancer Genome Atlas. There is an association between these two miRNAs and EOC, that was further verified by reverse transcription-quantitative polymerase chain reaction in peritoneal exosomes from 10 patients with EOC and NC participants. These results indicated that miR-149-3p and miR-222-5p might be novel biomarkers for evaluating the prognosis of patients with EOC and that these two miRNAs might have potential therapeutic values.

18.
Cancer Med ; 7(7): 3393-3405, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29797792

ABSTRACT

The therapeutic challenge of advanced, recurrent, and refractory cervical cancer (CC) needs to develop new molecularly targeted drugs. Rad21 is an important regulatory gene that maintains the correct dissociation of sister chromatids during cell mitosis. The aim of this study was to investigate the effect of Rad21 on CC. Rad21 expression in CC and cervical intraepithelial neoplasia III was significantly increased. Women with the rs2289937 C genotype (CC+CT) of rs4570 and rs4579555 genotypes and haplotype 1 (TTTCAGGCGC) were significantly associated with CC risk, while women with low frequencies of haplotype 6 (TTTTAGGCGC) also increased the risk of CC.Rad21-specific shRNA decreased cancerous cell proliferation, migration, and invasion and increased the proportion of cells in G2/M phase as well as sensitivity to radiation. The Rad21 influenced the expression of XPO1, CyclinB1, CDK1, P21, P27, and P53 through up-and downregulating the Rad21 expression. The TCGA database of CC also showed that Rad21 expression was associated with poor disease survival and XPO1 expression. Moreover, the KEGG pathway indicated that Rad21 is broadly involved in the cell cycle and RNA transportation via XPO1. This suggests that Rad21 involves the development of cervical cancer possibly by participating in the regulation of cell cycle and the nuclear output of the tumor suppressor gene via XPO1.

19.
Oncol Lett ; 13(5): 3219-3224, 2017 May.
Article in English | MEDLINE | ID: mdl-28521428

ABSTRACT

The aim of this study was to explore the expression of heparanase (HPA) in metastatic lymph nodes (LNs) of cervical cancer and to evaluate HPA as a marker of micro-metastasis of LNs. Immunohistochemistry was performed to detect the expression of HPA in 53 cases with metastasis of LNs (group A) and 49 cases without (group B). Scoring was determined based on the intensity of immunostaining and the size of the staining area. Three points or higher score was considered as positive. Among all cases, the positive rate of HPA was 76.5% in primary lesions and 84.9% in both primary lesions and metastatic LNs in group A. In group B, the rates were 67.3% in primary lesions and 8.2% in metastatic LNs. The expression of HPA in group A was significantly higher than that in group B (P<0.05). Compared with stage IA-IB and well-differentiated and non-metastatic LNs, the LNs of stage IIA and moderately/poorly differentiated and metastatic LNs expressed higher HPA (P<0.05). The overall 5-year survival rate was 73.3% and the median overall survival time (MOS) was 49.0 months. The MOS of the two groups was 36.0 and 58.5 months, respectively (P=0.023); the MOS of patients with positive HPA expression was distinctly lower than that of negative patients (P=0.040). Clinical staging, degree of differentiation, lymph node metastasis and expression of HPA notably affected patient prognosis; lymph node metastasis and expression of HPA were independent risk factors affecting patient prognosis (P<0.05). Our study demonstrated that high-level expression of HPA in cervical cancer was involved in LN metastasis, further impacting on patients' long-term survival. The clinical value of HPA requires further in-depth study.

20.
Medicine (Baltimore) ; 96(2): e5768, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28079802

ABSTRACT

BACKGROUND: DNA methylation modification has been proved to influence the phenotype of polycystic ovary syndrome (PCOS). Genome-wide association studies (GWAS) demonstrate that yes-associated protein (YAP1) genetic sites are associated with PCOS. The study aims to detect the methylation status of YAP1 promoter in ovary granulosa cells (GCs) of PCOS patients and explore novel therapeutic targets for PCOS. METHODS: Randomized controlled trial was applied and a total of 72 women were included in the study, including 36 cases of PCOS patients and 36 cases of health controls. Ovary GCs were extracted from in vitro fertilization embryo transfer. Methylation status of YAP1 promoter was detected by bisulfite sequencing PCR (BSP). Protein and mRNA expression of YAP1 were measured by western blotting and real-time quantitate PCR. RESULTS: Overall methylation level of YAP1 promoter region from PCOS group was significantly lower than that from control group. CpG sites analysis revealed that 12 sites (-443, -431, -403, -371, -331, -120, -49, -5, +1, +9, +15, +22) were significantly hypomethylated in women with PCOS (P < 0.05). A significant upregulation of YAP1 mRNA and protein expression levels was observed. Testosterone concentration could alleviate the methylation status and demonstrate obvious dose-dependent relation. CONCLUSION: Our research achievements manifest that hypomethylation of YAP1 promoter promotes the YAP1 expression, which plays a key role in the pathogenesis and accelerate PCOS.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Phosphoproteins/genetics , Polycystic Ovary Syndrome/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Case-Control Studies , DNA Methylation , Female , Follicle Stimulating Hormone , Granulosa Cells/metabolism , Humans , Luteinizing Hormone , Phosphoproteins/metabolism , Polycystic Ovary Syndrome/metabolism , Promoter Regions, Genetic , Testosterone , Transcription Factors , YAP-Signaling Proteins , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL