Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Discov Oncol ; 15(1): 141, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38696071

ABSTRACT

PURPOSE: Cuproptosis, programmed cell death by intracellular copper-mediated lipoylated protein aggregation, is involved in various tumorigenesis and drug resistance abilities by mediating the tumor microenvironment. Previous studies have demonstrated that serum copper levels are higher in OC patients than in normal subjects. However, the exact relationship between cuproptosis and ovarian cancer progression remains to be further elucidated. METHODS: The Cancer Genome Atlas (TCGA) and gene expression omnibus (GEO) datasets were utilized to establish a cuproptosis-related prognostic signature in ovarian cancer. Subsequently, the bulk RNA-seq analysis and single-cell RNA-seq analysis were used to identify the relationship between signature with immune cell infiltration, chemotherapy, and cuproptosis-related scoring (CuRS) system. Finally, the potential biological functional roles of target genes in cuproptosis were validated in vitro. RESULTS: By using LASSO-Cox regression analysis to establish the cuproptosis-related prognostic model, our works demonstrated the accuracy and efficiency of our model in the TCGA (583 OC patients) and GEO (260 OC patients) OC cohorts, and the high-scoring groups showed worse survival outcomes. Notably, there were substantial differences between the high and low-risk groups in extensive respects, such as the activating transcription factors, cell pseudotime features, cell intercommunication patterns, immunocytes infiltration, chemotherapy response, and potential drug resistance. KIF26B was selected to construct a prognostic model from the identified 33 prognosis-related genes, and high expression of KIF26B predicted poorer prognosis in ovarian cancer. Ultimately, further in vitro experiments demonstrated that KIF26B participated in the proliferation and cisplatin resistance of OC cells. Knockdown of KIF26B increased the sensitivity of OC cells to elesclomol, a cuproptosis agonists. CONCLUSION: This study constructed a new cuproptosis-related gene signature that has a good prognostic capacity in assessing the outcome of OC patients. This study enhances our understanding of cuproptosis associated with ovarian cancer aggressiveness, cross-talk with immunocytes, and serves as a novel chemotherapy strategy.

2.
Transl Oncol ; 44: 101917, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38554571

ABSTRACT

BACKGROUND: Single-cell sequencing was employed to analyze the tumor immune microenvironment in ovarian cancer (OC) patients, exploring the evolutionary roles of various macrophage subgroups in OC progression and their correlation with fatty acid metabolism-related genes in contributing to drug resistance. METHODS: This study aimed to decipher the mechanisms underlying OC chemoresistance (OC-CR) and carboplatin resistance by integrating and analyzing multiple single-cell RNA sequencing datasets from OC patients. The tumor immune microenvironment in OC-CR patients exhibited notable alterations in cellular interactions and the proportions of different immune cell populations, in contrast to the cohort sensitive to OC chemotherapy. RESULTS: The study demonstrates that the fatty acid-associated gene HEBP2 not only accelerates OC progression but also modifies the immune landscape of OC, driving the polarization from M0_TAM to M2_TAM. This shift results in a diminished efficacy of chemotherapy in OC. Furthermore, both in vitro and in vivo experiments underscored HEBP2's role in boosting the proliferation of OC-resistant cell lines and suppressing apoptosis, thereby facilitating carboplatin resistance. CONCLUSION: In conclusion, the immune microenvironments of OC-CR significantly differ from those sensitive to chemotherapy, underscoring HEBP2's role in fostering OC resistance. This establishes HEBP2 as a promising prognostic marker and a novel target for therapeutic strategies against OC resistance.

3.
Funct Integr Genomics ; 23(2): 147, 2023 May 05.
Article in English | MEDLINE | ID: mdl-37145301

ABSTRACT

Ovarian cancer (OC) has the worst prognosis among gynecological malignancies. Cisplatin (CDDP) is one of the most commonly used treatments for OC, but recurrence and metastasis are common due to endogenous or acquired resistance. High expression of ATP-binding cassette (ABC) transporters is an important mechanism of resistance to OC chemotherapy, but targeting ABC transporters in OC therapy remains a challenge. The expression of sortilin-related receptor 1 (SORL1; SorLA) in the response of OC to CDDP was determined by analysis of TCGA and GEO public datasets. Immunohistochemistry and western blotting were utilized to evaluate the expression levels of SORL1 in OC tissues and cells that were sensitive or resistant to CDDP treatment. The in vitro effect of SORL1 on OC cisplatin resistance was proven by CCK-8 and cell apoptosis assays. The subcutaneous xenotransplantation model verified the in vivo significance of SORL1 in OC. Finally, the molecular mechanism by which SORL1 regulates OC cisplatin resistance was revealed by coimmunoprecipitation, gene set enrichment analysis and immunofluorescence analysis. This study demonstrated that SORL1 is closely related to CDDP resistance and predicts a poor prognosis in OC. In vivo xenograft experiments showed that SORL1 knockdown significantly enhanced the effect of CDDP on CDDP-resistant OC cells. Mechanistically, silencing of SORL1 inhibits the early endosomal antigen 1 (EEA1) pathway, which impedes the stability of ATP-binding cassette B subfamily member 1 (ABCB1), sensitizing CDDP-resistant OC cells to CDDP. The findings of this study suggest that targeting SORL1 may represent a promising therapeutic approach for overcoming CDDP resistance in OC.


Subject(s)
Cisplatin , Ovarian Neoplasms , Humans , Female , Cisplatin/pharmacology , Cisplatin/therapeutic use , Cisplatin/metabolism , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphate , LDL-Receptor Related Proteins/metabolism , LDL-Receptor Related Proteins/pharmacology , LDL-Receptor Related Proteins/therapeutic use , Membrane Transport Proteins , ATP Binding Cassette Transporter, Subfamily B/pharmacology , ATP Binding Cassette Transporter, Subfamily B/therapeutic use
4.
Front Cell Dev Biol ; 9: 732786, 2021.
Article in English | MEDLINE | ID: mdl-34650978

ABSTRACT

Background: Importin 7 (IPO7), a karyopherin-ß protein, is involved in various tumorigenesis and progression abilities by mediating the nuclear import of oncoproteins. However, the exact biological functions of IPO7 remain to be further elucidated. Materials and Methods: TCGA and GEO datasets were used to identify dysregulated expression of IPO7 in various cancers. Gain-of-function and loss-of-function analyses were used to identify the oncogenic functions of IPO7 in vitro and in vivo. Moreover, LC-MS/MS and parallel reaction monitoring analysis were used to comparatively profiled IPO7-related proteomics and potential molecular machinery. Results: Our works demonstrated that the expression of IPO7 was upregulated and was correlated with a poor prognosis in cervical cancer. In vitro and in vivo experiments demonstrated that knockdown of IPO7 inhibited the proliferation of HeLa and C-4 I cells. LC-MS/MS analysis showed that IPO7-related cargo proteins mainly were enriched in gene transcription regulation. Then independent PRM analysis for the first time demonstrated that 32 novel IPO7 cargo proteins, such as GTF2I, RORC1, PSPC1, and RBM25. Moreover, IPO7 contributed to activating the PI3K/AKT-mTOR pathway by mediating the nuclear import of GTF2I in cervical cancer cells. Intriguingly, we found that the IPO7 expression was negatively correlated with CD8 T cell infiltration via regulating the expression of CD276 in cervical cancer. Conclusion: This study enhances our understanding of IPO7 nuclear-cytoplasmic translocation and might reveal novel potential therapeutic targets. The results of a negative correlation between the IPO7 and CD8 T cell infiltration indicate that the IPO7 might play an important impact on the immune microenvironment of cervical cancer.

5.
BMC Pregnancy Childbirth ; 21(1): 697, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34654394

ABSTRACT

BACKGROUND: Cesarean scar pregnancy (CSP) remains a sporadic and special form of ectopic pregnancy in which the fertilized ovum is implanted on a previous cesarean scar within 12 weeks. This study aims to evaluate the optimal time interval between uterine artery embolization (UAE) and curettage modalities in order to provide the best clinical outcomes. METHODS: From January 2018 to December 2020, we recruited 61 patients with CSP. They were randomly divided into two groups depending on whether the time interval between UAE and dilatation and curettage (D&C) requires additional hospitalization: 31 patients received prophylactic UAE followed by D&C on the same day (0-12 h; group A) and 30 patients need hospitalization (12-72 h; group B). The clinical characteristics, diagnostic data, and outcomes of the two groups were compared and analyzed. RESULTS: A total of 59 (96.72%) cases had responded well to the first treatment. One patient in each arm undergone retreatment, but none of the 61 patients needed additional hysterectomy. There was no considerable relationship between the two groups with respect to the intraoperative hemorrhage during D&C, serum index (containing ß-hCG, hemoglobin, CRP, and D-dimer) on the first day after D&C, side effects (containing fever and abdominal pain), renal, hepatic, and coagulation function, time of CSP residual mass disappearance, and hospitalization cost. The time of serum ß-hCG resolution after surgery was 41.22 ± 14.97 days in group A and 66.67 ± 36.64 days in group B (P = 0.027), and group A treatment resulted in a shorten hospital stay as compared with group B (4.81 ± 2.74 days vs. 6.80 ± 2.14 days, P <  0.001). However, the average hourly serum ß-hCG decrease rate within 24 h and the leukocytes on the first day after D&C in group B were superior than in group A (P <  0.050). CONCLUSION: For patients with CSP, UAE followed by D&C on the same day (0-12 h) appears to have more advantages in hospitalization and recovery time, while the long time interval (12-72 h) may have a lower risk of inflammation and a more rapid decrease in serum ß-hCG level within 24 h after D&C surgery. The treatment of CSP should be individualized based on the conditions of patients.


Subject(s)
Dilatation and Curettage/methods , Pregnancy, Ectopic/therapy , Uterine Artery Embolization/methods , Adult , Cesarean Section/adverse effects , China , Chorionic Gonadotropin, beta Subunit, Human/blood , Cicatrix/etiology , Female , Humans , Hysterectomy , Length of Stay , Pregnancy , Prospective Studies , Time Factors , Treatment Outcome
6.
Cancer Lett ; 515: 14-27, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34044070

ABSTRACT

Karyopherin-ß proteins are critically involved in cancer progression and have been reported as potential biomarkers and therapeutic targets for tumor treatment. However, TNPO1, as an important karyopherin-ß family member, underlying functional roles in cancers remain largely unclear. In this study, under integrated gene-expression profiling screen of karyopherin-ß in gynecologic cancer, we identify TNPO1 as a pivotal contributor to the gynecologic cancer progression. Remarkably, ARID1A-deficient gynecologic cancer cells are specifically vulnerable to the genetic perturbations of TNPO1 in vitro and in vivo. Mechanistically, TNPO1 is selectively responsible for nuclear import of ARID1B, which is a synthetic lethal target in ARID1A-inactivating mutation cancers. Furthermore, TNPO1 or ARID1B knockdown changes chromatin accessibility that results in loss of H3K4me1 and H3K27ac marker, diminishing activated transcription factor of the AP-1 family, and inactivating the PI3K/AKT signaling pathway by reducing growth pathway genes expression including PIK3CA and FGFR2. Together, this work indicates that the oncogenic function of TNPO1 and maybe represent a novel therapeutic strategy to treat ARID1A-deficient gynecologic cancer.


Subject(s)
Cell Proliferation/genetics , DNA-Binding Proteins/genetics , Genital Neoplasms, Female/genetics , Transcription Factors/genetics , beta Karyopherins/genetics , Animals , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Genital Neoplasms, Female/pathology , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation/genetics
7.
Cancer Lett ; 513: 36-49, 2021 08 10.
Article in English | MEDLINE | ID: mdl-33991616

ABSTRACT

Endometrial cancer (EC) is becoming one of the most common gynecologic malignancies. Lipid metabolism is a hallmark feature of cancers. The molecular mechanisms underlying lipid metabolism in EC remain unclear. In this study, we revealed that many lipid metabolism-related genes were aberrantly expressed in endometrial cancer tissues, especially ACLY. Upregulated ACLY promoted EC cell proliferation and colony formation, and attenuated apoptosis. Mechanistically, cotreatment with obesity-related factors (estradiol, insulin and leptin) promoted nuclear translocation of ACLY through Akt-mediated phosphorylation of ACLY at Ser455. Nuclear-localized ACLY increased histone acetylation levels, thus resulting in upregulation of pyrimidine metabolism genes, such as DHODH. Moreover, STAT3 altered the ACLY expression at the transcriptional level via directly binding to its promoter region. In conclusion, our findings clarify the roles and mechanisms of ACLY in endometrial cancer and ACLY could link obesity risk factors to the regulation of histone acetylation. We believe that novel therapeutic strategies for EC can be designed by targeting the ACLY axis.


Subject(s)
ATP Citrate (pro-S)-Lyase/metabolism , Endometrial Neoplasms/metabolism , Acetylation , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Endometrial Neoplasms/pathology , Female , HEK293 Cells , Heterografts , Humans , Lipid Metabolism , Mice , Middle Aged , Pyrimidines/metabolism , STAT3 Transcription Factor/metabolism
8.
J Immunol Res ; 2021: 9994004, 2021.
Article in English | MEDLINE | ID: mdl-33987449

ABSTRACT

Far upstream element binding protein 1 (FUBP1), a DNA-binding protein, participates in diverse tumor-promoting behaviors by regulating the expression of oncogenes in the nucleus, but the underlying mechanisms remain to be elucidated. In the present study, we found that FUBP1 mRNA and protein expressions were markedly upregulated and closely linked with poor prognosis in cervical cancer. In vitro, functional experiments showed that knockdown of FUBP1 inhibited CC cell proliferation and migration. Therefore, FUBP1 plays a prooncogenic function in CC progression. Further investigations for the first time demonstrated that nuclear localization of FUBP1 regulated the gene expression of immune checkpoint NRP1. Moreover, our work demonstrated that FUBP1 translocated into the nucleus which was mediated by interacting with Transportin-1 (TNPO1). Collectively, this study revealed that FUBP1 might be a potential therapeutic target for the restriction of tumor progression.


Subject(s)
DNA-Binding Proteins/metabolism , Neuropilin-1/genetics , RNA-Binding Proteins/metabolism , Tumor Escape/genetics , Uterine Cervical Neoplasms/immunology , beta Karyopherins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/immunology , Cell Nucleus/metabolism , Cell Proliferation/genetics , Cervix Uteri/pathology , DNA-Binding Proteins/genetics , Datasets as Topic , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/immunology , Gene Knockdown Techniques , HEK293 Cells , Humans , Kaplan-Meier Estimate , Prognosis , RNA-Binding Proteins/genetics , Tissue Array Analysis , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/mortality , Uterine Cervical Neoplasms/pathology
9.
J Immunol Res ; 2021: 6645885, 2021.
Article in English | MEDLINE | ID: mdl-33628849

ABSTRACT

Cervical cancer (CC) is a commonly diagnosed and primary consideration of cancer patient death in female reproductive system malignancy. Cyclin-dependent kinase 12 (CDK12), as a transcription-associated CDK, plays important roles in tumor-promoting behaviors, whereas the underlying mechanisms of CDK12 in CC progression are still obscure. In this report, we investigated the role of CDK12 in cervical cancer. The current study identified CDK12 mRNA and protein expression remarkably upregulated in CC patients. Upregulated CDK12 was closely associated with CC progression and poor prognosis. In vitro and in vivo functional experiments showed that knockdown of CDK12 inhibited cancer cell proliferation and colony formation and promoted apoptosis. Further investigations demonstrated that CDK12 regulated the immune microenvironment to facilitate the progression of CC cells by promoting macrophage infiltration. Meanwhile, we first demonstrated that nuclear import of CDK12 is mediated by TNPO1 and might be a new therapeutic target in oncology. Collectively, this study pointed out the potential of CDK12 to serve as a novel therapeutic target in restricting CC proliferation and cell cycle process through promoting macrophage infiltration.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Macrophages/immunology , Macrophages/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Animals , Apoptosis/genetics , Biomarkers , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinases/genetics , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Immunohistochemistry , Macrophages/pathology , Mice , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Prognosis , Protein Transport , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology , Uterine Cervical Neoplasms/mortality , Uterine Cervical Neoplasms/pathology , Xenograft Model Antitumor Assays
10.
Oncogene ; 39(34): 5633-5648, 2020 08.
Article in English | MEDLINE | ID: mdl-32661323

ABSTRACT

Cervical cancer (CC) remains highest in the mortality of female reproductive system cancers, while cisplatin (CDDP) resistance is the one of main reasons for the lethality. Preceding evidence has supported that karyopherins are associated with chemoresistance. In this study, we simultaneously compared CDDP-incomplete responders with CDDP-complete responders of CC patients and CDDP-insensitive CC cell lines with CDDP-sensitive group. We finally identified that DNA-PKcs (PRKDC) was related to CDDP sensitivity after overlapping in CC sample tissues and CC cell lines. Further functional assay revealed that targeting PRKDC by shRNA and NU7026 (specific PRKDC inhibitor) could enhance CDDP sensitivity in vitro and in vivo, which was mediated by impairing DNA damage repair pathway in CC. Mechanistically, we found that PRKDC was transcriptionally upregulated by CCAAT/enhancer-binding protein delta (CEBPD), while intriguingly, CDDP treatment strengthened the transcriptional activity of CEBPD to PRKDC. We further disclosed that Importin 4 (IPO4) augmented the nuclear translocation of CEBPD through nuclear localization signals (NLS) to activate PRKDC-mediated DNA damage repair in response to CDDP. Moreover, we demonstrated that IPO4 and CEBPD knockdown improved CDDP-induced cytotoxicity in vitro and in vivo. Together, we shed the novel insight into the role of IPO4 in chemosensitivity and provide a clinical translational potential to enhance CC chemosensitivity since the IPO4-CEBPD-PRKDC axis is actionable via NU7026 (PRKDC inhibitor) or targeting IPO4 in combination with CDDP.


Subject(s)
CCAAT-Enhancer-Binding Protein-delta/genetics , Cisplatin/pharmacology , DNA Repair/drug effects , DNA-Activated Protein Kinase/genetics , Membrane Transport Proteins/genetics , Uterine Cervical Neoplasms/genetics , Active Transport, Cell Nucleus/genetics , Animals , Antineoplastic Agents/pharmacology , CCAAT-Enhancer-Binding Protein-delta/metabolism , Cell Line, Tumor , Chromones/pharmacology , DNA Damage , DNA Repair/genetics , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/metabolism , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Membrane Transport Proteins/metabolism , Mice, Nude , Morpholines/pharmacology , RNA Interference , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/therapy , Xenograft Model Antitumor Assays/methods
11.
EBioMedicine ; 51: 102609, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31915116

ABSTRACT

BACKGROUND: Endometrial carcinoma (EC) is one of the most common gynecological malignancies among women. Maternal embryonic leucine Zipper Kinase (MELK) is upregulated in a variety of human tumors, where it contributes to malignant phenotype and correlates with a poor prognosis. However, the biological function of MELK in EC progression remains largely unknown. METHODS: We explored the MELK expression in EC using TCGA and GEO databases and verified it using clinical samples by IHC methods. CCK-8 assay, colony formation assay, cell cycle assay, wound healing assay and subcutaneous xenograft mouse model were generated to estimate the functions of MELK and its inhibitor OTSSP167. qRT-PCR, western blotting, co-immunoprecipitation, chromatin immunoprecipitation and luciferase reporter assay were performed to uncover the underlying mechanism concerning MELK during the progression of EC. FINDINGS: MELK was significantly elevated in patients with EC, and high expression of MELK was associated with serous EC, high histological grade, advanced clinical stage and reduced overall survival and disease-free survival. MELK knockdown decreased the ability of cell proliferation and migration in vitro and subcutaneous tumorigenesis in vivo. In addition, high expression of MELK could be regulated by transcription factor E2F1. Moreover, we found that MELK had a direct interaction with MLST8 and then activated mTORC1 and mTORC2 signaling pathway for EC progression. Furthermore, OTSSP167, an effective inhibitor, could inhibit cell proliferation driven by MELK in vivo and vitro assays. INTERPRETATION: We have explored the crucial role of the E2F1/MELK/mTORC1/2 axis in the progression of EC, which could be served as potential therapeutic targets for treatment of EC. FUNDING: This research was supported by National Natural Science Foundation of China (No:81672565), the Natural Science Foundation of Shanghai (Grant NO:17ZR1421400 to Dr. Zhihong Ai) and the fundamental research funds for central universities (No: 22120180595).


Subject(s)
Disease Progression , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cohort Studies , E2F1 Transcription Factor/metabolism , Endometrial Neoplasms/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Models, Biological , Multivariate Analysis , Naphthyridines/pharmacology , Prognosis , Protein Binding/drug effects , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , mTOR Associated Protein, LST8 Homolog/metabolism
12.
Biochem Biophys Res Commun ; 522(4): 952-959, 2020 02 19.
Article in English | MEDLINE | ID: mdl-31810606

ABSTRACT

Gallbladder carcinoma (GBC) is the most aggressive carcinoma of the biliary tract, effective chemotherapy was critical for the patients with unresectable GBC. However, chemotherapy resistance is still problematic for clinicians. Here, we identified a specific long non-coding RNA, SSTR5-AS1, in GBC patient that facilitates gemcitabine resistance. SSTR5-AS1 is significantly increased in GBC samples and cell lines, especially in gemcitabine-resistant cell lines, and higher SSTR5-AS1 expression was correlated with poorer overall survival rate in GBC patients. Our data revealed that upregulated SSTR5-AS1 facilitates gemcitabine resistance via inhibiting apoptosis. Knockdown of SSTR5-AS1 sensitized drug resistant GBC cells to gemcitabine in vitro and strongly inhibited xenografts formed by drug resistant GBC cells in vivo. Moreover, we found via streptavidin pull down assay that NONO specifically binds to sense sequence of SSTR5-AS1 and prevented proteasome mediated NONO degradation, which resulted in increased NONO protein level without affecting the transcription of NONO. NONO functions as the downstream effector of SSTR5-AS1 and is required for SSTR5-AS1 mediated gemcitabine resistance. Collectively, our data provided novel insights into lncRNA-mediated chemotherapy resistance and suggested a novel therapeutic target to improve chemotherapy strategies for unresectable GBC patients.


Subject(s)
DNA-Binding Proteins/metabolism , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , Gallbladder Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/drug effects , Humans , Mice, Nude , Proteasome Endopeptidase Complex/metabolism , Protein Stability/drug effects , Proteolysis/drug effects , RNA, Long Noncoding/genetics , Up-Regulation/drug effects , Up-Regulation/genetics , Gemcitabine
13.
J Cell Biochem ; 120(5): 8300-8310, 2019 May.
Article in English | MEDLINE | ID: mdl-30548294

ABSTRACT

PURPOSE: The dysregulation of long noncoding RNAs (lncRNAs) has been reported to be correlated to carcinogenesis and cancer progression. Endometrial cancer (EC), arising from the endometrium or the inner lining of the uterus, is one of the most common gynecological malignancies. We aim to explore the prognostic value of the lncRNAs in patients with endometrioid endometrial cancer (EEC) and to identify the potential lncRNA signature for predicting survival of patients with EEC. METHODS: We performed a genome-wide analysis of the lncRNA expression profiling in The Cancer Genome Atlas (TCGA)-Uterine Corpus Endometrial Carcinoma database (408 EEC) to identify the prognosis related lncRNAs for the EEC. The patients with EEC were randomly divided into a training set (204 for endometrioid) and a testing set (204 for endometrioid). The lncRNA signature was identified in the training set, and then independently validated in the testing set and the complete set (training set plus testing set). RESULTS: We developed a nine-lncRNA signature-based risk score in the patients with EEC. The risk score based on the novel lncRNAs signature was able to separate patients of training set into high-and low-risk groups with significantly different overall survival and progression-free survival. These can also be successfully confirmed in the testing set and complete set. CONCLUSION: A nine-lncRNA expression signature was identified and validated which can predict EEC patient's survival. These findings may have important implications in the understanding of the potential therapeutic methods for patients with EEC.

14.
Onco Targets Ther ; 10: 2951-2961, 2017.
Article in English | MEDLINE | ID: mdl-28652777

ABSTRACT

PURPOSE: Cancer stem cells have recently been identified as a key driving factor for tumor metastasis and chemoresistance. CD117 is a well-established cancer stem cell marker, but its clinical significance in epithelial ovarian cancer (EOC) remains controversial. Therefore, we aimed to identify correlations between CD117 expression and clinical features and outcomes in EOC patients in this meta-analysis. MATERIALS AND METHODS: A literature search was performed in the PubMed, Cochrane Library, Web of Science, EMBASE, and OVID databases to identify eligible studies. Correlations between CD117 expression and clinicopathological parameters and overall survival or disease-free survival were analyzed. A subgroup analysis was then performed, which was classified by patient ethnicity and age at diagnosis, study sample size, and tumor histological type. RESULTS: A total of seven studies enrolling 1,247 EOC patients were included in this meta-analysis. Our results demonstrated that CD117 expression was significantly correlated with age (pooled odds ratio [OR] =1.67, 95% confidence interval [CI] =1.05-2.66), International Federation of Gynecology and Obstetrics stage (pooled OR =1.99, 95% CI =1.31-3.02), tumor differentiation grade (pooled OR =2.46, 95% CI =1.48-4.10), and histological type (pooled OR =1.85, 95% CI =1.05-3.26). EOC patients with high CD117 expression had significantly worse OS (hazard ratio [HR] =1.39, 95% CI =1.03-1.90) than patients with low CD117 expression. However, no significant correlation was found between CD117 expression and disease-free survival (HR =1.31, 95% CI =0.79-2.17). In subgroup analysis, CD117 was identified as a significant prognostic factor for overall survival in European patients (HR =1.59, 95% CI =1.13-2.23), younger patients (<60 years) (HR =1.59, 95% CI =1.10-2.30), studies with sample sizes >200 (HR =1.84, 95% CI =1.32-2.56), and the mixed histological types (HR =1.47; 95% CI =1.08-2.00). CONCLUSION: Our meta-analysis suggests that CD117 is associated with EOC progression and can serve as a promising prognostic predictor for EOC patients. However, larger scale multicenter clinical trials are still needed to further validate our results.

SELECTION OF CITATIONS
SEARCH DETAIL
...