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










Publication year range
1.
Front Cell Dev Biol ; 12: 1384233, 2024.
Article in English | MEDLINE | ID: mdl-38872933

ABSTRACT

Background: Polycystic ovary syndrome (PCOS) is characterized by excess androgens, ovulatory dysfunction, and polycystic ovaries. The mechanisms underlying ovulatory and metabolic disorders in PCOS remain elusive, hampering therapeutic development. Enhanced metabolic health correlates with increased microbiota gene content and microbial diversity. We aimed to explore the impact of gut microbiota and serum steroids on PCOS regulation associated with androgen excess. Methods: The fecal samples of patients with hyperandrogenic PCOS (n = 14) and control group with PCOS (n = 14) were analyzed by 16S rRNA gene sequencing. The peripheral venous blood of all subjects was collected to detect serum hormones. The association between gut microbiota and serum hormones was analyzed with the R language. Results: Our findings reveal that the hyperandrogenic PCOS group exhibits lower richness and diversity of gut microbiota compared to the control group. Characteristic genera in PCOS patients with hyperandrogenism include Bifidobacterium, Enterobacteriaceae_unclassified, Streptococcus, Saccharimonadaceae, Enterococcus, and Eubacterium_nodatum_group. Five hormones, including 5ß-androsterone, deoxycorticosterone, corticosterone, 11-dehydrocorticosterone, and cortexolone, emerge as potential serum biomarkers for identifying patients with hyperandrogenic-PCOS (HA-PCOS). Furthermore, a lower vitamin D3 level may act as a susceptibility factor, suggesting that vitamin D3 supplementation could serve as a potential intervention for PCOS with hyperandrogenism. Conclusion: Specific fecal microbiota and serum steroids may be used as characteristic markers for clinical diagnosis of hyperandrogenic-PCOS. This research enhances our understanding of the intricate interplay among hormones, gut microbiota, and hyperandrogenemia in patients with PCOS.

2.
Protein Cell ; 15(6): 460-473, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38441496

ABSTRACT

The current coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus (SARS-CoV-2) remains a threat to pregnant women. However, the impact of early pregnancy SARS-CoV-2 infection on the maternal-fetal interface remains poorly understood. Here, we present a comprehensive analysis of single-cell transcriptomics and metabolomics in placental samples infected with SARS-CoV-2 during early pregnancy. Compared to control placentas, SARS-CoV-2 infection elicited immune responses at the maternal-fetal interface and induced metabolic alterations in amino acid and phospholipid profiles during the initial weeks post-infection. However, subsequent immune cell activation and heightened immune tolerance in trophoblast cells established a novel dynamic equilibrium that mitigated the impact on the maternal-fetal interface. Notably, the immune response and metabolic alterations at the maternal-fetal interface exhibited a gradual decline during the second trimester. Our study underscores the adaptive immune tolerance mechanisms and establishment of immunological balance during the first two trimesters following maternal SARS-CoV-2 infection.


Subject(s)
COVID-19 , Placenta , Pregnancy Complications, Infectious , SARS-CoV-2 , Female , Pregnancy , Humans , COVID-19/immunology , COVID-19/virology , SARS-CoV-2/immunology , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , Placenta/immunology , Placenta/virology , Placenta/metabolism , Immune Tolerance , Trophoblasts/immunology , Trophoblasts/metabolism , Trophoblasts/virology , Adult , Pregnancy Trimester, First/immunology , Transcriptome
3.
FASEB J ; 38(1): e23372, 2024 01.
Article in English | MEDLINE | ID: mdl-38102977

ABSTRACT

Embryo vitrification is a standard procedure in assisted reproductive technology. Previous studies have shown that frozen embryo transfer is associated with an elevated risk of adverse maternal and neonatal outcomes. This study aimed to explore the effects of mouse blastocyst vitrification on the phenotype of vitrified-warmed blastocysts, their intrauterine and postnatal development, and the long-term metabolic health of the derived offspring. The vitrified-warmed blastocysts (IVF + VT group) exhibited reduced mitochondrial activity, increased apoptotic levels, and decreased cell numbers when compared to the fresh blastocysts (IVF group). Implantation rates, live pup rates, and crown-rump length at E18.5 were not different between the two groups. However, there was a significant decrease in fetal weight and fetal/placental weight ratio in the IVF + VT group. Furthermore, the offspring of the IVF + VT group at an age of 36 weeks had reduced whole energy consumption, impaired glucose and lipid metabolism when compared with the IVF group. Notably, RNA-seq results unveiled disturbed hepatic gene expression in the offspring from vitrified-warmed blastocysts. This study revealed the short-term negative impacts of vitrification on embryo and fetal development and the long-term influence on glucose and lipid metabolism that persist from the prenatal stage into adulthood in mice.


Subject(s)
Cryopreservation , Vitrification , Pregnancy , Female , Animals , Mice , Cryopreservation/methods , Placenta , Embryonic Development , Blastocyst , Glucose , Retrospective Studies
4.
Oncogenesis ; 9(9): 81, 2020 Sep 11.
Article in English | MEDLINE | ID: mdl-32913192

ABSTRACT

Endometrial cancer stem cells (ECSCs) are stem-like cells endowed with self-renewal and differentiation abilities, and these cells are essential for cancer progression in endometrial cancer (EC). As hallmarks of the tumour microenvironment (TME), hypoxia and hypoxia-inducing factors (HIFs) give rise to the dysregulation of tumour stemness genes, such as SOX2. Against this backdrop, we investigated the regulatory mechanisms regulated by HIFs and SOX2 in ECSCs during EC development. Here, ECSCs isolated from EC cell lines and tissues were found to express stemness genes (CD133 and aldehyde dehydrogenase, ALDH1) following the induction of their ECSC expansion. Notably, m6A methylation of RNA and HIF-1α/2α-dependent AlkB homologue 5 (ALKBH5) participate in the regulation of HIFs and SOX2 in EC, as confirmed by the observations that mRNA levels of m6A demethylases and ALKBH5 significantly increase under hypoxic conditions in ECSCs. Moreover, hypoxia and high ALKBH5 levels restore the stem-like state of differentiated ECSCs and increase the ECSC-like phenotype, whereas the knockdown of HIFs or ALKBH5 significantly reduces their tumour initiation capacity. In addition, our findings validate the role of ALKBH5 in promoting SOX2 transcription via mRNA demethylation, thereby maintaining the stem-like state and tumorigenicity potential of ECSCs. In conclusion, these observations demonstrate a critical role for m6A methylation-mediated regulation of the HIF-ALKBH5-SOX2 axis during ECSC expansion in hypoxic TMEs.

5.
FASEB J ; 34(8): 10966-10983, 2020 08.
Article in English | MEDLINE | ID: mdl-32592217

ABSTRACT

Uterine leiomyomas (ULM) are a major public health issue contributing to high morbidity and poor pregnancy outcomes. However, its molecular pathogenesis is poorly understood. HMGA2-ULM is the second major subtype of human ULM and associates with large sizes, fast-growth, and high percentages of estrogen receptor α (ERα). As altered ERα expression plays a distinct role in ULM growth, here, we investigate a regulatory mechanism driving ULM growth via HMGA2 and ERα. We reveal a positive correlation of HMGA2 with ERα protein and demonstrate that HMGA2 promotes ULM cells proliferation via ERα. In addition, autophagy pathway and p62/SQSTM1 (a selective autophagy receptor) are found to participate in the regulation of HMGA2 and ERα. Moreover, HMGA2 suppresses the transcription of p62 by binding to its promoter, meanwhile, p62 interacts with ERα, and inhibition of p62 increases ERα expression and enhances cell viability in ULM, suggesting a novel mechanism of the HMGA2-p62-ERα axis in ULM proliferation. Notably, rapamycin, a familiar autophagy agonist, reduces ERα levels and the proliferation ability of ULM cells. This study demonstrates a causal role of the HMGA2-p62-ERα axis in preventing autophagy and increasing ERα expression in HMGA2-ULM. Therefore, blocking HMGA2-p62-ERα axis and targeting autophagy pathway establish a roadmap toward HMGA2-ULM medical treatment.


Subject(s)
Cell Proliferation/physiology , Estrogen Receptor alpha/genetics , HMGA2 Protein/genetics , Leiomyoma/genetics , Sequestosome-1 Protein/genetics , Uterine Neoplasms/genetics , Adult , Autophagy/physiology , Cell Survival/genetics , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Leiomyoma/pathology , Middle Aged , Promoter Regions, Genetic/genetics , Signal Transduction/genetics , Transcription, Genetic/genetics , Uterine Neoplasms/pathology , Uterus/pathology
6.
Biomed Pharmacother ; 129: 110427, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32574974

ABSTRACT

Triptolide is a multi-functional natural small molecular compound extracted from a traditional Chinese medicinal herb. Triptolide and its derivatives exhibit cytotoxicity through inducing DNA damage, therefore increasing sensitivity to DNA-damage based chemotherapy or radiotherapy in different types of cells. However, the regulatory mechanism of genotoxicity by triptolide, and the loss of genome integrity induced by triptolide are not fully understood. Here, we measured the effects of triptolide on genome integrity in a human fibroblast line HCA2-hTERT using the neutral comet assay. We demonstrated that treating cells with triptolide induced genomic instability in HCA2-hTERT cells. Furthermore, we observed the accumulation of γH2AX foci in triptolide treated cells than control cells at 24 h post ionizing radiation. Further mechanistic studies indicated that triptolide inhibited the enzymatic activity of DNA-PKcs, the critical nonhomologous end joining factor. In vitro kinase activity assays showed that triptolide suppressed the kinase activity of DNA-PKcs and molecular docking also predicted a potential interaction between triptolide and DNA-PKcs. As a consequence, we found that triptolide treatment enhanced the interaction between DNA-PKcs and KU80 and hampered the following recruitment of 53BP1. Altogether, our finding provides a new perspective about the toxicity of triptolide in non-cancer cells and highlights the necessity of taking genome effects of triptolide and its derivatives into consideration in the future clinical and research applications.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair/drug effects , DNA-Activated Protein Kinase/antagonists & inhibitors , Diterpenes/toxicity , Fibroblasts/drug effects , Genomic Instability/drug effects , Phenanthrenes/toxicity , Protein Kinase Inhibitors/pharmacology , Cell Line , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , Epoxy Compounds/toxicity , Fibroblasts/enzymology , Fibroblasts/pathology , Histones/metabolism , Humans , Ku Autoantigen/metabolism , Phosphorylation , Telomerase/genetics , Telomerase/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism
7.
Signal Transduct Target Ther ; 5(1): 81, 2020 05 27.
Article in English | MEDLINE | ID: mdl-32457342

ABSTRACT

The Rho family of GTPases is strictly regulated by a large family of GTPase-activating proteins (GAPs) that stimulate the relatively weak intrinsic GTP-hydrolyzing activity of Rho GTPases. p190A is a potent and widely expressed GAP that acts on RhoA GTPases. p190A is frequently mutated in endometrial cancer, but the contribution of p190A mutations to endometrial tumorigenesis remains unclear. Here we identified that p190A is an upstream regulator of the Hippo-YAP signaling pathway, which is a critical regulator of cell proliferation, apoptosis, and cell fate. p190A knockout in endometrial cancer cells promoted cell proliferation, migration, and epithelial-mesenchymal transition (EMT), which were partially dependent on YAP activation. Wild-type p190A, but not endometrial cancer-associated mutants, suppressed the nuclear localization, transcriptional activity, and malignant transformation function of YAP. Moreover, the nuclear localization of YAP was enhanced in p190A-mutated endometrial cancer. These findings reveal novel molecular mechanisms underlying Hippo-YAP pathway-driven endometrial tumorigenesis and elucidate the potential for therapy targeting the Hippo-YAP pathway in p190A-mutated endometrial cancer.


Subject(s)
Cell Cycle Proteins/genetics , Endometrial Neoplasms/genetics , Guanine Nucleotide Exchange Factors/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , rhoA GTP-Binding Protein/genetics , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Hippo Signaling Pathway , Humans , Mutation/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics
8.
Onco Targets Ther ; 12: 2235-2245, 2019.
Article in English | MEDLINE | ID: mdl-30988628

ABSTRACT

OBJECTIVE: The aim of this study was to examine the effect of TRIB3 on proliferation, apoptosis, and migration of endometrial cancer (EC) cells and explore the relationship between TRIB3 and AKT signaling pathway in EC progression. METHODS: Immunohistochemical analysis was performed to measure the expression level of TRIB3 in normal endometrium tissues and EC tissues. Overexpression and shRNA knockdown techniques were applied by transfecting EC cells (ISK and AN3CA), and the effect of TRIB3 on EC cell biological behaviors was evaluated. Cell Counting Kit-8 and colony formation assays were utilized to investigate EC cell proliferation ability, and flow cytometry was performed to assess the apoptosis of EC cells. Moreover, the migration and invasion of EC cells were detected by transwell assay, and the levels of MMP-2 and MMP-9 were measured by ELISA. Additionally, Western blot analysis was carried out to determine the levels of AKT and p-AKT. RESULTS: The expression level of TRIB3 was higher in EC than normal endometrium tissues, and its overexpression promoted apoptosis and suppressed proliferation of EC cells. Furthermore, TRIB3 retarded the migration and invasion of EC cells and decreased the levels of MMP-2 and MMP-9. Conversely, TRIB3 inhibition enhanced the expression levels of MMP-2 and MMP-9, and proliferation and migration of EC cells but suppressed their apoptosis. Similarly, TRIB3 overexpression reduced while its knockdown increased the level of p-AKT. CONCLUSION: TRIB3 inhibited proliferation and migration and promoted apoptosis of EC cells probably through regulating AKT signaling pathway.

9.
Oncotarget ; 8(43): 74479-74493, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-29088801

ABSTRACT

FAM83D, a microtubule-associated protein (MAP), is overexpressed in diverse types of human cancer. The expression and critical role of FAM83D in human gastric cancer (GC), however, remains largely unknown. Here, we conducted molecular, cellular and clinical analyses to evaluate the functional link of FAM83D to GC. FAM83D expression was elevated in gastric tumors, and its expression strongly correlated with lymph node metastasis and TNM stage. In addition, over-expression of FAM83D in GC cell lines enhanced cell proliferation, cycle progression, migration, invasion, as well as tumor growth and metastatic dissemination in vivo. Furthermore, FAM83D exhibited a strong cell cycle correlated expression. The knockdown of FAM83D inhibited the regrowth of microtubules in GC cells. FAM83D was co-immunoprecipitated with HMMR, TPX2, and AURKA, a set of drivers of mitosis progression. Taken together, our results demonstrate FAM83D as an important player in the development of human gastric cancer, and as a potential therapeutic target for the treatment of cancer.

10.
PLoS One ; 11(4): e0152228, 2016.
Article in English | MEDLINE | ID: mdl-27050168

ABSTRACT

Recent research on bilateral transfer suggests that imagery training can facilitate the transfer of motor skill from a trained limb to that of an untrained limb above and beyond that of physical practice. To further explore this effect, the present study examined the influence of practice duration and task difficulty on the extent to which imagery training and physical training influences bilateral transfer of a sequential key pressing task. In experiment 1, participants trained on the key pressing task using their non-dominant arm under one of three conditions (physical practice, imagery practice, and no practice). In a subsequent bilateral transfer test, participants performed the sequential task using their untrained dominant arm in either an original order or mirror-ordered sequence. In experiment 2, the same procedures were followed as in experiment 1 except that participants trained with their dominant arm and performed the bilateral transfer task with their non-dominant arm. Results indicated that with extended practice beyond what has been employed in previous studies, physical practice is more effective at facilitating bilateral transfer compared to training with imagery. Interestingly, significant bilateral transfer was only observed for transfer from the non-dominant to the dominant arm with no differences observed between performing the task in an original or mirror ordered sequence. Overall, these findings suggest that imagery training may benefit bilateral transfer primarily at the initial stages of learning, but with extended training, physical practice leads to larger influences on transfer.


Subject(s)
Imagery, Psychotherapy , Motor Skills , Muscle, Skeletal/physiology , Psychomotor Performance , Adult , Electromyography , Humans , Young Adult
11.
Anticancer Agents Med Chem ; 16(9): 1085-92, 2016.
Article in English | MEDLINE | ID: mdl-27025656

ABSTRACT

It has been suggested that administration of the omega-3 polyunsaturated fatty acids (ω-3 PUFAs), including docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), can alter the toxicity and/or activity of several anticancer drugs in in vitro and in vivo studies. Here, we investigated the ability of ω-3 PUFAs to potentiate the antineoplastic activity of cisplatin (CDDP) in gastric cancer cells. The increase in CDDP-induced growth inhibition was measured by the IC50 values obtained when the cells were incubated with CDDP alone or with CDDP plus DHA or EPA. DHA and EPA enhanced the growth-inhibition activity of increasing concentrations of CDDP. The interactions between CDDP and DHA or EPA at the cellular level were assessed through the combination index (CI) method of Chou-Talalay. The results demonstrated synergism between CDDP and DHA or EPA in MKN45 cells. Cell cycle analysis showed that the combination treatment increased G0/G1 phase and S phase arrest, and significantly increased the number of apoptotic cells. According to our previous study, ω -3 PUFAs induce apoptosis of gastric cells via ADORA1, a subtype of adenosine receptor functionally related to cell death. The ADORA1 mRNA and protein expression was higher in the combination treatment than in the individual treatments. Notable, when GC cells were pretreated with DPCPX, a selective ADORA1 antagonist, the combination treatment effect on apoptosis was significantly reduced. Our results suggest that ω-3 PUFAs enhance the antineoplastic effects of CDDP in gastric cancer cells, and the synergistic effect between ω-3 PUFAs and CDDP is partly dependent on activating the ADORA1-mediated apoptosis pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cisplatin/pharmacology , Fatty Acids, Omega-3/pharmacology , Receptor, Adenosine A1/metabolism , Stomach Neoplasms/drug therapy , Cell Cycle/drug effects , Cell Line, Tumor , Drug Synergism , Gastric Mucosa/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , RNA, Messenger/genetics , Receptor, Adenosine A1/genetics , Stomach/drug effects , Stomach/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism
12.
Cancer Lett ; 360(2): 269-79, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25697482

ABSTRACT

P53 mutation plays a pivotal role in tumorigenesis of endometrial cancer (EC), here we report that the gain-of-function mutant p53-R248Q targets the proteasome activator REGγ to promote EC progression. Increased p53 expression significantly correlated with high pathological grade and lymph node metastasis in EC specimens. Manipulation of p53-R248Q in EC cells caused coincident changes in REGγ expression, and chromatin immunoprecipitation coupled with PCR further indicated that p53-R248Q bound to the REGγ gene promoter at a p53 responsive element. Silencing of REGγ in EC cells attenuated the cell proliferation, migration and invasion abilities, whereas overexpression of p53-R248Q rescued these activities. Overexpression of REGγ also induced an epithelial-mesenchymal transition phenotype. Moreover, a mouse xenograft tumor model showed that REGγ promoted tumor growth, further demonstrating a p53-R248Q-REGγ oncogenic pathway. Finally, examination of EC and normal endometrium specimens confirmed the oncogenic role of REGγ, in that REGγ was more highly overexpressed in p53-positive specimens than in p53-negative specimens. Our data suggest that REGγ is a promising therapeutic target for EC with the p53-R248Q mutation.


Subject(s)
Autoantigens/genetics , Carcinoma, Endometrioid/genetics , Endometrial Neoplasms/genetics , Genes, p53 , Mutation , Proteasome Endopeptidase Complex/genetics , Animals , Autoantigens/biosynthesis , Carcinoma, Endometrioid/enzymology , Cell Growth Processes/genetics , Cell Movement/genetics , Endometrial Neoplasms/enzymology , Epithelial-Mesenchymal Transition , Female , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Paraffin Embedding , Proteasome Endopeptidase Complex/biosynthesis , Signal Transduction , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Up-Regulation
13.
Front Biosci (Landmark Ed) ; 19(6): 854-61, 2014 06 01.
Article in English | MEDLINE | ID: mdl-24896321

ABSTRACT

Omega-3 polyunsaturated fatty acids (Omega-3 PUFAs), including docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), have been suggested to have anti-cancer effects by epidemiological and clinical studies. However, their underlying anti-cancer mechanisms are still unclear. In this study, we examined the influence of two Omega-3 PUFAs (DHA and EPA) on the proliferation and apoptosis of gastric cancer (GC) cells, and found that DHA and EPA reduced the viability of GC cells and induced apoptosis by activating caspase-3. Moreover, we screened the expression profile of apoptosis-related genes in GC cells upon the treatment of DHA and/or EPA, and discovered that ADORA1, one subtype of adenosine receptor functionally involved in cell death, was up-regulated in response to DHA and EPA. Importantly, when GC cells were treated with a selective ADORA1 antagonist, DPCPX, the DHA/EPA-induced apoptosis was substantially reduced. Taken together, our results suggest that the anti-cancer effect of Omega-3 PUFAs on gastric cancer is at least partly dependent on activating the ADORA1-mediated apoptosis pathway.


Subject(s)
Apoptosis/drug effects , Apoptosis/genetics , Fatty Acids, Omega-3/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Receptor, Adenosine A1/genetics , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Docosahexaenoic Acids/pharmacology , Dose-Response Relationship, Drug , Eicosapentaenoic Acid/pharmacology , Flow Cytometry , Humans , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Time Factors
14.
Biochem Biophys Res Commun ; 446(1): 167-72, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24582558

ABSTRACT

Interleukin (IL)-6 as an inflammation factor, has been proved to promote cancer proliferation in several human cancers. However, its role in endometrial cancer has not been studied clearly. Previously, we demonstrated that IL-6 promoted endometrial cancer progression through local estrogen biosynthesis. In this study, we proved that IL-6 could directly stimulate endometrial cancer cells proliferation and an autocrine feedback loop increased its production even after the withdrawal of IL-6 from the medium. Next, we analyzed the mechanism underlying IL-6 production in the feedback loop and found that its production and IL-6-stimulated cell proliferation were effectively blocked by pharmacologic inhibitors of nuclear factor-kappa B (NF-κB) and extra-cellular signal-regulated kinase (ERK). Importantly, activation of ERK was upstream of the NF-κB pathways, revealing the hierarchy of this event. Finally, we used an orthotopic nude endometrial carcinoma model to confirm the effects of IL-6 on the tumor progression. Taken together, these data indicate that IL-6 promotes endometrial carcinoma growth through an expanded autocrine regulatory loop and implicate the ERK-NF-κB pathway as a critical mediator of IL-6 production, implying IL-6 to be an important therapeutic target in endometrial carcinoma.


Subject(s)
Endometrial Neoplasms/metabolism , Interleukin-6/metabolism , MAP Kinase Signaling System , NF-kappa B/metabolism , Animals , Autocrine Communication , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation , Endometrial Neoplasms/etiology , Endometrial Neoplasms/pathology , Feedback, Physiological , Female , Heterografts , Humans , Interleukin-6/genetics , Mice , Mice, Nude , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Signal Transduction
15.
PLoS One ; 9(1): e87563, 2014.
Article in English | MEDLINE | ID: mdl-24498137

ABSTRACT

MicroRNAs play key roles in tumor proliferation and invasion. Here we show distinct expression of miR-222-3p between ERα-positive and ERα-negative endometrial carcinoma (EC) cell lines and primary tumors, and investigation of its relationship with ERα and other clinical parameters. In vitro, the function of miR-222-3p was examined in RL95-2 and AN3CA cell lines. MiR-222-3p expression was negatively correlated with ERα. Over-expressed miR-222-3p in RL95-2 cells promoted cell proliferation, enhanced invasiveness and induced a G1 to S phase shift in cell cycle. Furthermore, the miR-222-3p inhibitor decreased the activity of AN3CA cells to proliferate and invade. In vivo, down-regulated miR-222-3p of AN3CA cells inhibited EC tumor growth in a mouse xenograft model. Additionally, miR-222-3p increased raloxifene resistance through suppressing ERα expression in EC cells. In conclusion, miR-222-3p plays a significant role in the regulation of ERα expression and could be potential targets for restoring ERα expression and responding to antiestrogen therapy in a subset of ECs.


Subject(s)
Endometrial Neoplasms/metabolism , Estrogen Receptor alpha/biosynthesis , G1 Phase , MicroRNAs/biosynthesis , Neoplasm Proteins/biosynthesis , RNA, Neoplasm/biosynthesis , S Phase , Adult , Animals , Cell Line, Tumor , Endometrial Neoplasms/pathology , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , MicroRNAs/genetics , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/genetics , RNA, Neoplasm/genetics
16.
Int J Cancer ; 135(2): 282-94, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24347287

ABSTRACT

Tumor-stroma interactions contribute greatly to intratumoral estrogen biosynthesis in endometrial carcinoma, but the mechanisms involved remain largely unknown. Previous study demonstrated that intratumoral aromatase upregulation in stromal cells participated in this process, but the specific aromatase-regulators have not been reported. In the present study, we found that aromatase expression in intratumoral stroma, but not in tumor epithelium, correlated positively with interleukin 6 (IL-6) expression in cancer epithelial cells by immunohistochemistry, which was confirmed using laser capture microdissection/real-time reverse transcription-PCR. With stimulation by exogenous IL-6, aromarase expression was increased in stromal cells not but not in cancer cells. Aromatase mRNA levels in endometrial cancer cells were not influenced by cocultivation with intratumoral stromal cells. When cocultured with 17ß-estradiol (E2 )-treated cancer cells, aromatase mRNA in stromal cells was significantly elevated and increased IL-6 protein levels were detected in E2 -treated culture medium. Next, we demonstrated that E2 -induced IL-6 production was through cooperation between estrogen receptor α and nuclear factor-kappa B. Furthermore, an IL-6 receptor blocking antibody could attenuate the upregulation of aromatase expression in stromal cells and the E2 concentration in coculture systems of cancer and stromal cells. The results were confirmed by an orthotopic nude endometrial carcinoma model in vivo. These studies elucidated the activation of a positive feedback loop, that is, IL-6 stimulated by E2 in endometrial cancer cells induced aromatase expression in stromal cells, promoting enhanced intratumoral E2 synthesis. Blocking of this tumor-stroma interaction may be a therapeutic strategy to overcome in situ estrogen biosynthesis in endometrial carcinoma.


Subject(s)
Aromatase/metabolism , Endometrial Neoplasms/metabolism , Estradiol/biosynthesis , Interleukin-6/metabolism , Tumor Microenvironment/physiology , Animals , Blotting, Western , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Feedback, Physiological , Female , Humans , Immunohistochemistry , Immunoprecipitation , Laser Capture Microdissection , Mice , Mice, Nude , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/metabolism
17.
Zhonghua Wei Chang Wai Ke Za Zhi ; 10(1): 70-2, 2007 Jan.
Article in Chinese | MEDLINE | ID: mdl-17253180

ABSTRACT

OBJECTIVE: To identify the expression of polo like kinase 1 (plk1) and to discuss its relationship with the clinicopathological parameters and prognosis in gastric carcinoma. METHODS: Plk1 protein expression levels in 89 cases of resected gastric carcinomas were detected by immunohistochemistry method, the relations between plk1 expression levels and the survival periods were estimated by Kaplan-Meier curve. RESULTS: The positive rate of plk1 expression in gastric cancer tissues was 42.7% (38/89), significantly higher than that (13.5%) in the adjacent noncancerous tissues (12/89) (P<0.01). The expression levels of plk1 were closely related to tumor differentiation, invasion and TNM stage (P<0.05). Patients with plk1-positive expression had worse prognosis than those with plk1-negative expression in gastric cancer patients (P<0.05). CONCLUSIONS: Plk1 may promote carcinogenesis and gastric cancer development, its overexpression can be a novel marker for diagnosing certain biological behaviours and predicting prognosis in gastric cancer.


Subject(s)
Cell Cycle Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Aged , Cell Cycle Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Male , Neoplasm Staging , Prognosis , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Stomach Neoplasms/genetics , Polo-Like Kinase 1
18.
Zhonghua Wei Chang Wai Ke Za Zhi ; 9(1): 62-6, 2006 Jan.
Article in Chinese | MEDLINE | ID: mdl-16437376

ABSTRACT

OBJECTIVE: To observe the effect of inhibition of polo like kinase1 (plk1) gene expression on apoptosis induction in gastric cancer cell line-MKN45 and discuss the vital role of plk1 proliferation and viability of gastric cancer cells. METHODS: The plk1 expression was inhibited by chemically synthesized siRNA. The plk1 mRNA and protein level were respectively measured by real-time quantitative PCR and Western blotting. The spindle morphological change was observed by immunofluorescence staining and confocal microscopy. The change of cell cycle distribution and apoptosis rate was detected by flow-cytometry. Pro caspase3 level was also detected by western blotting. RESULTS: After treatment by siRNA targeting plk1, plk1 mRNA and protein level decreased obviously, the cell mitotic spindle became obscure and lost cohesiveness, more MKN45 cells accumulated at G(2)/M phase (P< 0.05), apoptosis rate of plk1 siRNA treated MKN45 cells was higher than that of control cells at 48 h and 72 h (P< 0.05) with pro-caspase3 level decreasing at 72 h. CONCLUSIONS: Inhibition of plk1 gene expression induces apoptosis in MKN45 cells through the pathway of caspase3. Plk1 gene play a key role in viability of MKN45 cells.


Subject(s)
Apoptosis , Cell Cycle Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , Stomach Neoplasms/genetics , Cell Cycle , Cell Line, Tumor , Gene Expression , Humans , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Polo-Like Kinase 1
19.
Zhonghua Zhong Liu Za Zhi ; 28(8): 568-71, 2006 Aug.
Article in Chinese | MEDLINE | ID: mdl-17236548

ABSTRACT

OBJECTIVE: To detect the gene expression profile in gastric cancer cell cycle and explain the mechanism of gastric cancer cell proliferation by a genomic study. METHODS: Gastric cancer cells MKN45 were synchronized at G2/M and G1/S point by nocodazole-thymidine and double thymidine methods. The synchronizing degree of cells was monitored by flow cytometry. The gene expression profiles at G2/M point, M/G1 transition, G1 early phase, G1 late phase, G1/S point, S early phase, S late phase, G2 early phase and G2 late phase in MKN45 cell cycling were examined using cDNA microarray chips. Hierarchy analysis was conducted with a professional software package and the up-regulated genes at G1 late and G2 phase were analyzed according to gene database. Furthermore, the mRNA level of cyclin E, cyclin B, plk1 and STK15 in above mentioned nine points were measured by quatitative PCR. RESULTS: 2001 genes were detected to be available at all 9 points via software processing, out of which 959 appeared up-regulated or down-regulated. 379 genes showed to be up-regulated at late G1 (147) or G2 phases (232), 40 at S and M phases (also up-regulated at G1 late and G2 phases). The 147 up-regulated genes at G1 late phase are involved in DNA metabolism, transcription and translation, protein transportation, ubiquitination and signal transduction, etc. The 232 up-regulated genes in G2 phase are involved in RNA synthesis and processing, intracellular protein transportation, cytoskeleton synthesis, signal transduction, apoptosis and anti-apoptosis, transcription regulation, ubiquitination, mitosis regulation and oncogene expression, etc. The mRNA level of 4 genes detected by quantitative PCR during cell cycle was in agreement with that detected by microarray. CONCLUSION: During MKN45 cell cycling, the preparation for DNA synthesis and chromosome separation are conducted in G1 and G2, which are implicated in multiple genes, may be the main impetus of driving MKN45 cell cycle. Some of these genes may be related to tumor over-proliferation. The cDNA microarray technique has characteristic features such as reliability and can provide a great deal for future research on cell cycle related genes in gastric cancer.


Subject(s)
Cell Cycle/genetics , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis/methods , Aurora Kinase A , Aurora Kinases , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cyclin B/genetics , Cyclin E/genetics , Gene Expression Regulation, Neoplastic , Humans , Polymerase Chain Reaction/methods , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Polo-Like Kinase 1
20.
World J Gastroenterol ; 8(3): 436-40, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12046065

ABSTRACT

AIM: To investigate the effects of antisense human telomerase RNA (hTR)on the biologic behavior of human gastric cancer cell line: MKN-45 by gene transfection and its potential role in the gene therapy of gastric cancer. METHODS: The hTR cDNA fragment was cloned from MKN-45 through RT-PCR and subcloned into eukaryotic expression vector (pEF6/V5-His-TOPO) in cis-direction or trans-direction by DNA recombinant methods. The constructed sense, antisense and empty vectors were transfected into MKN-45 cell lines separately by lipofectin-mediated DNA transfection technology. After drug selection, the expression of antisense hTR gene in stable transfectants and normal MKN-45 cells was detected by RT-PCR, the telomerase activity by TRAP, the apoptotic features by PI and Hoechst 33258 staining, the cell cycle distribution by flow cytometry and the population doubling time by cell counting. Comparison among the stable transfectants and normal MKN-45 cells was made. RESULTS: The sense, antisense hTR eukaryotic expression vectors and empty vector were successfully constructed and proved to be the same as original design by restriction endonuclease analysis and sequencing. Then, they were successfully transfected into MKN-45 cell lines separately with lipofectin. The expression of antisense hTR gene was only detected in MKN-45 cells stably transfected with antisense hTR vector (named as MKN-45-ahTR) but not in the control cells. In MKN-45-ahTR, the telomerase activity was inhibited by 75%, the apoptotic rate was increased to 25.3%, the percentage of cells in the G0/G1 phase was increased to 65%, the proliferation index was decreased to 35% and the population doubling time was prolonged to 35.3 hours. However, the telomerase activity, the apoptotic rate, the distribution of cell cycle, the proliferation index and the population doubling time were not different among the control cells. CONCLUSION: Antisense hTR can significantly inhibit telomerase activity and proliferation of MKN-45 cells and induce cell apoptosis. Antisense gene therapy based on telomerase inhibition can be a potential therapeutic approach to the treatment of gastric cancer.


Subject(s)
RNA, Antisense/pharmacology , Telomerase/antagonists & inhibitors , Apoptosis/drug effects , Cell Division/drug effects , Gene Expression , Genetic Vectors , Humans , RNA, Antisense/genetics , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Stomach Neoplasms/therapy , Telomerase/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...