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










Publication year range
1.
Heliyon ; 9(6): e17493, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37408896

ABSTRACT

Purpose: To explore the mechanisms by which abnormal female BMI affects oocyte quality, particularly whether it involves the alteration of gene expression patterns and how these patterns may impact clinical outcomes. Methods: In Part 1, we performed a retrospective study to compare the clinical outcomes between the female BMI ≥25 kg/m2 and female BMI ≤20 kg/m2 groups. In Part 2, we performed the transcriptome analyses based on the GSE87201 dataset. Results: In Part 1, among the clinical outcomes, only the grade 1-2 embryo rate at day 3 of ICSI cycles was significantly different between the two BMI groups; the other outcomes were not. In Part 2, compared with the BMI ≤20 kg/m2 group, the oocyte gene expression pattern of the BMI ≥25 kg/m2 group seemed to result in better oocyte tolerance to exogenous stress, such as intracytoplasmic sperm injection (ICSI). It seemed to explain the result of Part 1 that the BMI ≥25 kg/m2 group had better day-3 embryo quality after ICSI than the BMI ≤20 kg/m2 group. Conclusions: Abnormal female BMI affects oocyte quality by altering the gene expression patterns of oocytes. While a female BMI ≥25 kg/m2 is known to have certain detrimental effects on ART, our findings suggest that it can also confer some benefits to oocytes.

2.
Syst Biol Reprod Med ; 69(3): 245-254, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36772853

ABSTRACT

Artificial oocyte activation (AOA) is considered an effective method to improve clinical outcomes in patients with some forms of male factor infertility and does not increase the risk of birth defects. However, the effects of AOA on patients with multiple morphological abnormalities of the sperm flagella (MMAF) caused by a DNAH1 mutation are still unknown. To explore the effects, our study analyzed a case with MMAF due to DNAH1 homozygous mutation that underwent testicular sperm extraction (TESE) combined with intracytoplasmic sperm injection (ICSI). The case had 28 MII oocytes. The 28 oocytes were divided randomly and equally into AOA and non-AOA groups. Ionomycin was used for AOA. We compared the clinical outcomes of two groups and selected three blastulation failure embryos from each group for transcriptome analysis (Data can be accessed through GSE216618). Differentially expressed genes (DEGs) were determined with an adjusted p-value <0.05 and a |log2-fold change| ≥1. The comparison of clinical outcomes showed that the two pronuclei (2PN) rate and grade 1-2 embryo rate at day 3 were not significantly different between the two groups. Transcriptome analyses of blastulation failed embryos showed that the use of AOA had potential risks of chromosome structure defects, transcriptional regulation defects, and epigenetic defects. In conclusion, when the case with MMAF due to DNAH1 mutation underwent TESE-ICSI, ionomycin-induced oocyte activation could not improve the clinical outcomes and introduced the risks of chromosome structure defect, transcriptional regulation defect, and epigenetic defect.


Subject(s)
Infertility, Male , Semen , Female , Humans , Male , Pregnancy , Flagella , Infertility, Male/genetics , Ionomycin/pharmacology , Oocytes , Pregnancy Rate , Spermatozoa
3.
Cancer Lett ; 524: 161-171, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34687791

ABSTRACT

Sorafenib and its derivative regorafenib are the first- and second-line targeted drugs for advanced HCC, respectively. Although both drugs improve overall survival, drug resistance remains the major barrier to their full efficacy. Thus, strategies to enhance sorafenib and regorafenib efficacy against HCC are solely needed. Interleukin-6 receptor alpha (IL-6Rα) is the receptor of IL-6, a multi-functional cytokine, which plays key roles in liver-regeneration, inflammation and development of hepatocellular carcinoma (HCC). Here we show the expression of IL-6Rα was induced in response to sorafenib. Depletion of IL-6Rα abolished IL-6 induced STAT3 phosphorylation at 705th tyrosine and tumor growth of HCC cells under sorafenib treatment. Mechanistically, activating transcription factor 3 (ATF3) was induced in response to sorafenib and subsequently bound to the promoter of IL-6Rα, leading to its transcriptional activation. Depletion of ATF3 or its upstream transcription factor, ATF4, attenuated IL-6Rα induction and IL-6 mediated sorafenib resistance. The ATF4-ATF3-IL-6Rα cascade is also activated by regorafenib. Furthermore, blockade of IL-6Rα with the FDA approved IL-6Rα antibody drug, Sarilumab, drastically attenuated both sorafenib and regorafenib resistance in patient-derived xenograft (PDX) tumors, where human IL-6 could be detected by a novel in situ hybridization technique, named RNAscope. Together, our data reveal that ATF3-mediated IL-6Rα up-regulation promotes both sorafenib and regorafenib resistance in HCC, and targeting IL-6Rα represents a novel therapeutic strategy to enhance sorafenib/regorafenib efficacy for advanced HCC treatment.


Subject(s)
Activating Transcription Factor 3/genetics , Carcinoma, Hepatocellular/drug therapy , Interleukin-6/genetics , Liver Neoplasms/drug therapy , Receptors, Interleukin-6/genetics , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Sorafenib/pharmacology , Xenograft Model Antitumor Assays
4.
Reprod Fertil Dev ; 33(18): 881-885, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34852900

ABSTRACT

Context The timing of early cumulus cell removal (ECCR) can be changed within a range. The change has an effect on the multiple pronuclei (MPN) rate and the exposure time of oocytes to sperm waste products. The timing of ECCR effects the outcomes of assisted reproductive technology, however, it is still unclear what time is best for ECCR. Aims To find the best time for ECCR based on clinical outcomes in order to increase the success rate of assisted reproductive technology. Methods A retrospective study was performed. Cycles were categorised into six groups according to the timing of ECCR. The clinical outcomes of these six groups were compared by Kruskal-Wallis test and Pearson X 2 test. Key results The timing of ECCR had a significant effect on the MPN rate, 0PN without cleavage rate and grade 1-2 embryo rate at Day3. Among our six time groups of ECCR, the cumulus cell removal ≤4h post-insemination group had the highest MPN rate and grade 1-2 embryo rate at Day3, and the 5.5h

Subject(s)
Cumulus Cells/physiology , Fertilization/physiology , Insemination, Artificial , Reproductive Techniques, Assisted , Embryo Transfer , Female , Humans , Male , Ovulation Induction , Pregnancy , Pregnancy Rate , Retrospective Studies , Sperm Injections, Intracytoplasmic
5.
Mol Carcinog ; 60(2): 151-163, 2021 02.
Article in English | MEDLINE | ID: mdl-33428809

ABSTRACT

Regorafenib is approved for patients with unresectable hepatocellular carcinoma (HCC) following sorafenib. However, the effect of regorafenib on HCC metastasis and its mechanism are poorly understood. Here, our data showed that regorafenib significantly restrained the migration, invasion and vasculogenic mimicry (VM) of HCC cells, and downregulated the expression of epithelial-to-mesenchymal transition (EMT)/VM-related molecules. Using RNA-seq and cellular thermal shift assays, we found that inhibitor of differentiation 1 (ID1) was a key target of regorafenib. In HCC tissues, the protein expression of ID1 was positively correlated with EMT and VM formation (CD34- /PAS+ ). Functionally, ID1 knockdown inhibited HCC cell migration, invasion, metastasis, and VM formation in vitro and in vivo, with upregulation of E-cadherin and downregulation of Snail and VE-cadherin. Moreover, Snail overexpression promoted the migration, invasion, and VM formation of ID1 knockdown cells. Snail knockdown reduced the migration, invasion, and VM formation of ID1 overexpression cells. Finally, regorafenib suppressed VM formation and decreased the expression of ID1, VE-cadherin and Snail in HCC PDX model. In conclusion, we manifested that regorafenib distinctly inhibited EMT in HCC cells via targeting ID1, leading to the suppression of cell migration, invasion and VM formation. These findings suggest that regorafenib may be developed as a suitable therapeutic agent for HCC metastasis.


Subject(s)
Carcinoma, Hepatocellular/prevention & control , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Liver Neoplasms/prevention & control , Neovascularization, Pathologic/prevention & control , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Xenograft Model Antitumor Assays/methods , Animals , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Liver Neoplasms/blood supply , Liver Neoplasms/genetics , Male , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neovascularization, Pathologic/genetics , Tumor Burden/drug effects , Tumor Burden/genetics
6.
J Assist Reprod Genet ; 37(3): 619-627, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31901111

ABSTRACT

OBJECTIVE: To investigate the role of the cell number at day 3 in blastocyst selection. DESIGN: Observational, retrospective, single-center clinical study. PATIENT(S): In part 1, 1211 single vitrified-warmed blastocyst transfer (SVBT) cycles were identified and reviewed. All the cycles were conventional in vitro fertilization (IVF) cycles and the first embryo transfer cycles. Most of patients had a risk of ovarian hyperstimulation syndrome and were young. In part 2, 864 IVF-derived blastocysts from 292 infertile couples underwent trophectoderm (TE) biopsy for preimplantation genetic testing for aneuploidies (PGT-A). INTERVENTION(S): No patient intervention. MAIN OUTCOME MEASURE(S): The first part was an analysis of the correlation between the cell number at day 3 and live birth rate (LBR) after SVBT, and the second part was an analysis of the correlation between the cell number at day 3 and euploid rate (ER) of blastocysts. RESULT(S): In part 1, after correcting for the effects of other confounders, the cell number at day 3 had no significant effect on the LBR (OR 1.001, 95% CI 0.938-1.068). In part 2, after correcting for the effects of other confounders, the cell number at day 3 had no significant effect on the ER (OR 0.960, 95% CI 0.866-1.063). CONCLUSION(S): When the vitrified-warmed blastocysts obtained by conventional IVF are transferred into young patients, the cell number at day 3 is not a strong predictor of the LBR. In addition, the cell number at day 3 is not a strong predictor of ER of IVF-derived blastocysts too.


Subject(s)
Blastocyst/cytology , Embryo Transfer , Embryonic Development/genetics , Preimplantation Diagnosis , Adult , Birth Rate , Cryopreservation , Embryo Implantation/genetics , Female , Fertilization in Vitro , Genetic Testing , Humans , Infertility/genetics , Infertility/pathology , Live Birth/genetics , Pregnancy
7.
Cancer Sci ; 110(8): 2442-2455, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31148345

ABSTRACT

The human prolyl isomerase PIN1, best known for its association with carcinogenesis, has recently been indicated in the disease of pancreatic ductal adenocarcinoma (PDAC). However, the functions of PIN1 and the feasibility of targeting PIN1 in PDAC remain elusive. For this purpose, we examined the expression of PIN1 in cancer, related paracarcinoma and metastatic cancer tissues by immunohistochemistry and analyzed the associations with the pathogenesis of PDAC in 173 patients. The functional roles of PIN1 in PDAC were explored in vitro and in vivo using both genetic and chemical PIN1 inhibition. We showed that PIN1 was upregulated in pancreatic cancer and metastatic tissues. High PIN1 expression is significantly association with poor clinicopathological features and shorter overall survival and disease-free survival. Further stratified analysis showed that PIN1 phenotypes refined prognostication in PDAC. Inhibition of PIN1 expression with RNA interference or with all trans retinoic acid decreased not only the growth but also the migration and invasion of PDAC cells through regulating the key molecules of multiple cancer-driving pathways, simultaneously resulting in cell cycle arrest and mesenchymal-epithelial transition in vitro. Furthermore, genetic and chemical PIN1 ablation showed dramatic inhibition of the tumorigenesis and metastatic spread and then reduced the tumor burden in vivo. We provided further evidence for the use of PIN1 as a promising therapeutic target in PDAC. Genetic and chemical PIN1 ablation exerted potent antitumor effects through blocking multiple cancer-driving pathways in PDAC. More potent and specific PIN1 targeted inhibitors could be exploited to treat this aggressive cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Neoplasm Metastasis/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Animals , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Pancreatic Ductal/pathology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Disease-Free Survival , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Metastasis/pathology , Pancreatic Neoplasms/pathology , Up-Regulation/drug effects , Up-Regulation/genetics
8.
Nat Immunol ; 16(11): 1142-52, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26414765

ABSTRACT

Mitochondria need to be juxtaposed to phagosomes for the synergistic production of ample reactive oxygen species (ROS) in phagocytes to kill pathogens. However, how phagosomes transmit signals to recruit mitochondria has remained unclear. Here we found that the kinases Mst1 and Mst2 functioned to control ROS production by regulating mitochondrial trafficking and mitochondrion-phagosome juxtaposition. Mst1 and Mst2 activated the GTPase Rac to promote Toll-like receptor (TLR)-triggered assembly of the TRAF6-ECSIT complex that is required for the recruitment of mitochondria to phagosomes. Inactive forms of Rac, including the human Rac2(D57N) mutant, disrupted the TRAF6-ECSIT complex by sequestering TRAF6 and substantially diminished ROS production and enhanced susceptibility to bacterial infection. Our findings demonstrate that the TLR-Mst1-Mst2-Rac signaling axis is critical for effective phagosome-mitochondrion function and bactericidal activity.


Subject(s)
Phagocytes/immunology , Phagocytes/metabolism , Protein Serine-Threonine Kinases/metabolism , Reactive Oxygen Species/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bacterial Infections/etiology , Bacterial Infections/immunology , Bacterial Infections/metabolism , Blood Bactericidal Activity/immunology , Cell Line , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , Mitochondria/immunology , Mitochondria/metabolism , Mitochondria/microbiology , Phagocytes/microbiology , Phagosomes/immunology , Phagosomes/metabolism , Phagosomes/microbiology , Protein Kinase C-alpha/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Sepsis/etiology , Sepsis/immunology , Sepsis/metabolism , Serine-Threonine Kinase 3 , Signal Transduction , TNF Receptor-Associated Factor 6 , Toll-Like Receptors/metabolism , Ubiquitination , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism
9.
Nat Commun ; 6: 6239, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25695629

ABSTRACT

The role of the unfolded protein response (UPR) in tissue homeostasis remains largely unknown. Here we find that loss of Mst1/2, the mammalian Hippo orthologues, or their regulator WW45, leads to a remarkably enlarged endoplasmic reticulum (ER) size-associated UPR. Intriguingly, attenuation of the UPR by tauroursodeoxycholic acid (TUDCA) diminishes Mst1/2 mutant-driven liver overgrowth and tumorigenesis by promoting nuclear exit and degradation of Hippo downstream effector Yap. Yap is required for UPR activity and ER expansion to alleviate ER stress. During the adaptive stage of the UPR, PERK kinase-eIF2α axis activates Yap, while prolonged ER stress-induced Hippo signalling triggers assembly of the GADD34/PP1 complex in a negative feedback loop to inhibit Yap and promote apoptosis. Significantly, the deregulation of UPR signals associated with Yap activation is found in a substantial fraction of human hepatocellular carcinoma (HCC). Thus, we conclude Yap integrates Hippo and UPR signalling to control liver size and tumorigenesis.


Subject(s)
Carcinogenesis/pathology , Liver Neoplasms/pathology , Liver/growth & development , Liver/pathology , Protein Serine-Threonine Kinases/metabolism , Taurochenodeoxycholic Acid/pharmacology , Unfolded Protein Response , Activating Transcription Factor 6/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blotting, Western , Carcinogenesis/drug effects , Cell Cycle Proteins , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Hep G2 Cells , Hepatocyte Growth Factor/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Hepatocytes/ultrastructure , Humans , Liver Neoplasms/enzymology , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Molecular Sequence Data , Mutation/genetics , Organ Size/drug effects , Phosphoproteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins/metabolism , Serine-Threonine Kinase 3 , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Unfolded Protein Response/drug effects , Unfolded Protein Response/genetics , YAP-Signaling Proteins
10.
Acta Biochim Biophys Sin (Shanghai) ; 47(1): 46-52, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25476204

ABSTRACT

The Hippo signaling pathway is an evolutionarily conserved signaling module that plays critical roles in liver size control and tumorigenesis. The Hippo pathway consists of a core kinase cascade in which the mammalian Ste20-like kinases (Mst1/2, orthologs of Drosophila Hippo) and their cofactor Salvador (Sav1) form a complex to phosphorylate and activate the large tumor suppressor (Lats1/2). Lats1/2 kinases in turn phosphorylate and inhibit the transcription co-activators, the Yes-associated protein (YAP) and the transcriptional co-activator with PDZ-binding motif (TAZ), two major downstream effectors of the Hippo pathway. Losses of the Hippo pathway components induce aberrant hepatomegaly and tumorigenesis, in which YAP coordinates regulation of cell proliferation and apoptosis and plays an essential role. This review summarizes the current findings of the regulation of Hippo signaling in liver regeneration and tumorigenesis, focusing on how the loss of tumor suppressor components of the Hippo pathway results in liver cancers and discussing the molecular mechanisms that regulate the expression and activation of its downstream effector YAP in liver tumorigenesis.


Subject(s)
Carcinogenesis , Liver Neoplasms/physiopathology , Liver Regeneration/physiology , Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis/physiology , Cell Proliferation/physiology , Drosophila Proteins/metabolism , Gene Expression Regulation/physiology , Hippo Signaling Pathway , Homeostasis/physiology , Humans , Liver Neoplasms/pathology , Models, Biological , Nuclear Proteins/metabolism , Organ Size/physiology , Signal Transduction/physiology , Trans-Activators/metabolism , Transcriptional Activation/physiology , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL