Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Medicine (Baltimore) ; 103(23): e38526, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38847685

ABSTRACT

Tubal pregnancy is a common cause of maternal mortality in early pregnancy. Transumbilical laparoendoscopic single-site surgery (TU-LESS) has gained popularity due to its safety and aesthetic advantages. However, the lack of affordable disposable entry platforms hinders its widespread adoption. This study aimed to investigate the learning curve of tubal pregnancy removal using single-incision multiport (SIMP) laparoscopy and provide guidance for novice gynecologists. A retrospective analysis was conducted on cases of ectopic pregnancy (EP) diagnosed at Dongguan Songshan Lake Central Hospital from June 2020 to June 2022. The analysis included 50 cases, with 25 undergoing single-port laparoscopy and 25 undergoing conventional laparoscopy (CL). Various indicators, including body mass index (BMI), previous pregnancies, mass size, hemoglobin levels, surgical duration, and complications, were collected. Learning curve analysis using the cumulative sum (CUSUM) technique was performed to assess procedural proficiency. There were no significant differences in patient characteristics or complications between the 2 groups. However, the single-port laparoscopy group exhibited a statistically significant longer average surgical time (41.60 ±â€…13.38 minutes) compared to the conventional laparotomy group (32.96 ±â€…7.32 minutes). The CUSUM analysis demonstrated a decline in surgical time after the completion of approximately 11 cases, indicating an improvement in SIMP laparoscopy surgical proficiency. SIMP laparoscopy for tubal pregnancy removal achieved similar safety outcomes as CL. Notably, the CUSUM analysis revealed that proficiency in single-port laparoscopy could be achieved after approximately 11 cases, leading to stable surgical times. These findings serve as valuable guidance for novice gynecologists interested in adopting single-incision laparoscopy.


Subject(s)
Laparoscopy , Learning Curve , Operative Time , Salpingectomy , Humans , Female , Retrospective Studies , Laparoscopy/methods , Laparoscopy/education , Salpingectomy/methods , Salpingectomy/education , Adult , Pregnancy , Pregnancy, Tubal/surgery , Clinical Competence
2.
Oncogene ; 42(47): 3491-3502, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37828220

ABSTRACT

Cell senescence deters the activation of various oncogenes. Induction of senescence is, therefore, a potentially effective strategy to interfere with vital processes in tumor cells. Sphingosine-1-phosphate receptor 1 (S1PR1) has been implicated in various cancer types, including ovarian cancer. The mechanism by which S1PR1 regulates ovarian cancer cell senescence is currently elusive. In this study, we demonstrate that S1PR1 was highly expressed in human ovarian cancer tissues and cell lines. S1PR1 deletion inhibited the proliferation and migration of ovarian cancer cells. S1PR1 deletion promoted ovarian cancer cell senescence and sensitized ovarian cancer cells to cisplatin chemotherapy. Exposure of ovarian cancer cells to sphingosine-1-phosphate (S1P) increased the expression of 3-phosphatidylinositol-dependent protein kinase 1 (PDK1), decreased the expression of large tumor suppressor 1/2 (LATS1/2), and induced phosphorylation of Yes-associated protein (p-YAP). Opposite results were obtained in S1PR1 knockout cells following pharmacological inhibition. After silencing LATS1/2 in S1PR1-deficient ovarian cancer cells, senescence was suppressed and S1PR1 expression was increased concomitantly with YAP expression. Transcriptional regulation of S1PR1 by YAP was confirmed by chromatin immunoprecipitation. Accordingly, the S1PR1-PDK1-LATS1/2-YAP pathway regulates ovarian cancer cell senescence and does so through a YAP-mediated feedback loop. S1PR1 constitutes a druggable target for the induction of senescence in ovarian cancer cells. Pharmacological intervention in the S1PR1-PDK1-LATS1/2-YAP signaling axis may augment the efficacy of standard chemotherapy.


Subject(s)
Ovarian Neoplasms , Protein Kinases , Female , Humans , Sphingosine-1-Phosphate Receptors/genetics , Ovarian Neoplasms/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Cellular Senescence/genetics , Cell Proliferation/genetics
3.
Epigenetics ; 18(1): 2175565, 2023 12.
Article in English | MEDLINE | ID: mdl-36908025

ABSTRACT

Platinum-based chemotherapy is one of the predominant strategies for treating ovarian cancer (OC), however, platinum resistance greatly influences the therapeutic effect. Circular RNAs (circRNAs) have been found to participate in the pathogenesis of platinum resistance. Our aim was to explore the involvement of circ_0078607 in OC cell cisplatin (DDP) resistance and its potential mechanisms. Circ_0078607, miR-196b-5p, and growth arrest-specific 7 (GAS7) levels were assessed by qPCR. Circ_0078607 stability was assessed by ribonuclease R digestion and actinomycin D treatment. Cell viability of various conic of DDP treatment was measured by CCK-8. The cell proliferation was determined by CCK-8 and colony formation assay. Western blotting was performed for determining GAS7, ABCB1, CyclinD1 and Bcl-2 protein levels. The direct binding between miR-196b-5p and circ_0078607 or GAS7 was validated by dual-luciferase reporter and RIP assay. DDP resistance in vivo was evaluated in nude mice. Immunohistochemistry staining for detecting Ki67 expression in xenograft tumours. Circ_0078607 and GAS7 was down-regulated, but miR-196b-5p was up-regulated in OC samples and DDP-resistant cells. Overexpression of circ_0078607 inhibited DDP resistance, cell growth and induced apoptosis in DDP-resistant OC cells. Mechanistically, circ_0078607 sequestered miR-196b-5p to up-regulate GAS7. MiR-196b-5p mimics reversed circ_0078607 or GAS7 overexpression-mediated enhanced sensitivity. Finally, circ_0078607 improved the sensitivity of DDP in vivo. Circ_0078607 attenuates DDP resistance via miR-196b-5p/GAS7 axis, which highlights the therapeutic potential of circ_0078607 to counter DDP resistance in OC.


Subject(s)
MicroRNAs , Nerve Tissue Proteins , Ovarian Neoplasms , Platinum , RNA, Circular , Animals , Female , Humans , Mice , Cell Proliferation , Cisplatin , DNA Methylation , Drug Resistance, Neoplasm , Mice, Nude , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Platinum/pharmacology , RNA, Circular/genetics
4.
Cancer Gene Ther ; 29(12): 1895-1907, 2022 12.
Article in English | MEDLINE | ID: mdl-35864225

ABSTRACT

Epigenetic alterations have been functionally linked to ovarian cancer development and occurrence. The CXXC zinc finger protein 1 (CFP1) is an epigenetic regulator involved in DNA methylation and histone modification in mammalian cells. However, its role in ovarian cancer cells is unknown. Here, we show that CFP1 protein is highly expressed in human ovarian cancer tissues. Loss of CFP1 inhibited the growth of human ovarian cancer cells, promoted apoptosis, and increased senescence. CFP1 knockdown resulted in reduced levels of SETD1 (a CFP1 partner) and histone H3 trimethylation at the fourth lysine residue (H3K4me3). RNA-sequencing revealed that deletion of CFP1 resulted in mRNA reduction of bone marrow stromal cell antigen 2 (BST2). Bioinformatics analysis and chromatin immunoprecipitation showed that CFP1 binds to the promoter of BST2 and regulates its transcription directly. Overexpression of BST2 rescued the growth inhibitory effect of CFP1 loss. Furthermore, depletion of cullin-RING ubiquitin ligases 4 (CRL4) components ROC1 or CUL4A had significantly inhibited the expression of CFP1 and BST2 similar to MLN4924 treatment that blocked cullin neddylation and inactivated CRL4s. In conclusion, CFP1 promotes ovarian cancer cell proliferation and apoptosis by regulating the transcription of BST2, and the expression of CFP1 was affected by CRL4 ubiquitin ligase complex.


Subject(s)
Antigens, CD , Ovarian Neoplasms , Trans-Activators , Female , Humans , Antigens, CD/genetics , Cell Proliferation/genetics , Cullin Proteins , GPI-Linked Proteins/genetics , Ovarian Neoplasms/genetics , Trans-Activators/genetics , Ubiquitins
6.
Nat Commun ; 13(1): 3034, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35641493

ABSTRACT

Abnormal neddylation activation is frequently observed in human cancers and neddylation inhibition has been proposed as a therapy for cancer. Here, we report that MLN4924, a small-molecule inhibitor of neddylation activating enzyme, increases glutamine uptake in breast cancer cells by causing accumulation of glutamine transporter ASCT2/SLC1A5, via inactivation of CRL3-SPOP E3 ligase. We show the E3 ligase SPOP promotes ASCT2 ubiquitylation, whereas SPOP itself is auto-ubiquitylated upon glutamine deprivation. Thus, SPOP and ASCT2 inversely regulate glutamine uptake and metabolism. SPOP knockdown increases ASCT2 levels to promote growth which is rescued by ASCT2 knockdown. Adding ASCT2 inhibitor V-9302 enhances MLN4924 suppression of tumor growth. In human breast cancer specimens, SPOP and ASCT2 levels are inversely correlated, whereas lower SPOP with higher ASCT2 predicts a worse patient survival. Collectively, our study links neddylation to glutamine metabolism via the SPOP-ASCT2 axis and provides a rational drug combination for enhanced cancer therapy.


Subject(s)
Breast Neoplasms , Nuclear Proteins , Repressor Proteins , Ubiquitin-Protein Ligases , Amino Acid Transport System ASC/genetics , Amino Acid Transport System ASC/metabolism , Cell Line, Tumor , Female , Glutamine/metabolism , Humans , Minor Histocompatibility Antigens/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
7.
Clin Transl Med ; 12(1): e703, 2022 01.
Article in English | MEDLINE | ID: mdl-35073459

ABSTRACT

BACKGROUND: Chemoresistance to cisplatin (DDP) remains a major challenge in advanced gastric cancer (GC) treatment. Although accumulating evidence suggests an association between dysregulation of long non-coding RNAs (lncRNAs) and chemoresistance, the regulatory functions and complexities of lncRNAs in modulating DDP-based chemotherapy in GC remain under-investigated. This study was designed to explore the critical chemoresistance-related lncRNAs in GC and identify novel therapeutic targets for patients with chemoresistant GC. METHODS: Chemoresistance-related lncRNAs were identified through microarray and verified through a quantitative real-time polymerase chain reaction (qRT-PCR). Proteins bound by lncRNAs were identified through a human proteome array and validated through RNA immunoprecipitation (RIP) and RNA pull-down assays. Co-immunoprecipitation and ubiquitination assays were performed to explore the molecular mechanisms of the Musashi2 (MSI2) post-modification. The effects of LINC00942 (LNC942) and MSI2 on DDP-based chemotherapy were investigated through MTS, apoptosis assays and xenograft tumour formation in vivo. RESULTS: LNC942 was found to be up-regulated in chemoresistant GC cells, and its high expression was positively correlated with the poor prognosis of patients with GC. Functional studies indicated that LNC942 confers chemoresistance to GC cells by impairing apoptosis and inducing stemness. Mechanically, LNC942 up-regulated the MSI2 expression by preventing its interaction with SCFß-TRCP E3 ubiquitin ligase, eventually inhibiting ubiquitination. Then, LNC942 stabilized c-Myc mRNA in an N6-methyladenosine (m6 A)-dependent manner. As a potential m6 A recognition protein, MSI2 stabilized c-Myc mRNA with m6 A modifications. Moreover, inhibition of the LNC942-MSI2-c-Myc axis was found to restore chemosensitivity both in vitro and in vivo. CONCLUSIONS: These results uncover a chemoresistant accelerating function of LNC942 in GC, and disrupting the LNC942-MSI2-c-Myc axis could be a novel therapeutic strategy for GC patients undergoing chemoresistance.


Subject(s)
Cisplatin/metabolism , Drug Resistance/drug effects , Genes, myc/drug effects , RNA, Long Noncoding/agonists , RNA-Binding Proteins/antagonists & inhibitors , Cisplatin/therapeutic use , Genes, myc/physiology , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/therapeutic use , RNA-Binding Proteins/genetics , RNA-Binding Proteins/pharmacology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics
8.
Clin Transl Med ; 11(10): e587, 2021 10.
Article in English | MEDLINE | ID: mdl-34709767

ABSTRACT

Chemoresistance remains a major obstacle to successful cancer therapy, especially for advanced cancers. It used to be recognised as a stable outcome resulting from genetic changes. However, recent studies showed that chemoresistance can also be unstable and reversible with the involvement of non-genetic alterations. In the present study, we found that activating transcription factor 4 (ATF4) is downregulated in chemoresistant gastric cancer cells. The over-expression of ATF4 reversed chemoresistance by activating CHOP transcription to enhance drug-induced apoptosis, and vice versa. Moreover, casein kinase 1 delta (CK1δ) was identified as the kinase responsible for ATF4-S219 phosphorylation, which triggered ßTrCP-mediated ATF4 polyubiquitination to promote its proteasomal degradation subsequently. Interestingly, drug withdrawal gradually restored chemosensitivity as well as ATF4 expression in chemoresistant cells, highlighting the dependence of dynamic drug resistance on ATF4 protein expression. In line with these findings, the inhibition of ATF4 protein degradation by CK1δ or proteasome inhibitors overcame chemoresistance both in vitro and in vivo. Taken together, these results indicate that CK1δ stimulates ßTrCP-dependent ATF4 polyubiquitination and subsequent proteasomal degradation to promote chemoresistance in gastric cancer. Stabilisation of the ATF4 protein with bortezomib (BTZ), an anticancer drug that inhibits proteasomal degradation, might be a rational strategy to improve chemotherapeutic efficacy in gastric cancer.


Subject(s)
Activating Transcription Factor 4/genetics , Casein Kinase Idelta/genetics , Casein Kinase Idelta/metabolism , Drug Resistance, Neoplasm/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Ubiquitination/genetics , Activating Transcription Factor 4/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Proteasome Endopeptidase Complex
9.
Cell Death Dis ; 12(4): 405, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33854041

ABSTRACT

p62/SQSTM1 is frequently up-regulated in many cancers including hepatocellular carcinoma. Highly expressed p62 promotes hepato-carcinogenesis by activating many signaling pathways including Nrf2, mTORC1, and NFκB signaling. However, the underlying mechanism for p62 up-regulation in hepatocellular carcinoma remains largely unclear. Herein, we confirmed that p62 was up-regulated in hepatocellular carcinoma and its higher expression was associated with shorter overall survival in patients. The knockdown of p62 in hepatocellular carcinoma cells decreased cell growth in vitro and in vivo. Intriguingly, p62 protein stability could be reduced by its acetylation at lysine 295, which was regulated by deacetylase Sirt1 and acetyltransferase GCN5. Acetylated p62 increased its association with the E3 ligase Keap1, which facilitated its poly-ubiquitination-dependent proteasomal degradation. Moreover, Sirt1 was up-regulated to deacetylate and stabilize p62 in hepatocellular carcinoma. Additionally, Hepatocyte Sirt1 conditional knockout mice developed much fewer liver tumors after Diethynitrosamine treatment, which could be reversed by the re-introduction of exogenous p62. Taken together, Sirt1 deacetylates p62 at lysine 295 to disturb Keap1-mediated p62 poly-ubiquitination, thus up-regulating p62 expression to promote hepato-carcinogenesis. Therefore, targeting Sirt1 or p62 is a reasonable strategy for the treatment of hepatocellular carcinoma.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Sirtuin 1/metabolism , Animals , Autophagy/physiology , Kelch-Like ECH-Associated Protein 1/metabolism , Mice, Inbred BALB C , Protein Processing, Post-Translational/physiology , Signal Transduction/physiology
10.
Cell Biosci ; 11(1): 55, 2021 Mar 17.
Article in English | MEDLINE | ID: mdl-33731189

ABSTRACT

Mitochondria are the powerhouse of a cell. The structure and function of mitochondria are precisely regulated by multiple signaling pathways. Neddylation, a post-translational modification, plays a crucial role in various cellular processes including cellular metabolism via modulating the activity, function and subcellular localization of its substrates. Recently, accumulated data demonstrated that neddylation is involved in regulation of morphology, trafficking and function of mitochondria. Mechanistic elucidation of how mitochondria is modulated by neddylation would further our understanding of mitochondrial regulation to a new level. In this review, we first briefly introduce mitochondria, then neddylation cascade, and known protein substrates subjected to neddylation modification. Next, we summarize current available data of how neddylation enzymes, its substrates (including cullins/Cullin-RING E3 ligases and non-cullins) and its inhibitor MLN4924 regulate the structure and function of mitochondria. Finally, we propose the future perspectives on this emerging and exciting field of mitochondrial research.

11.
Front Cell Dev Biol ; 9: 741736, 2021.
Article in English | MEDLINE | ID: mdl-34977006

ABSTRACT

Hypoxia is an important characteristic of the tumor microenvironment. Tumor cells can survive and propagate under the hypoxia stress by activating a series of adaption response. Herein, we found that lysine-specific demethylase 5B (KDM5B) was upregulated in gastric cancer (GC) under hypoxia conditions. The genetic knockdown or chemical inhibition of KDM5B impaired the growth of GC cell adapted to hypoxia. Interestingly, the upregulation of KDM5B in hypoxia response was associated with the SUMOylation of KDM5B. SUMOylation stabilized KDM5B protein by reducing the competitive modification of ubiquitination. Furthermore, the protein inhibitor of activated STAT 4 (PIAS4) was determined as the SUMO E3 ligase, showing increased interaction with KDM5B under hypoxia conditions. The inhibition of KDM5B caused significant downregulation of hypoxia-inducible factor-1α (HIF-1α) protein and target genes under hypoxia. As a result, co-targeting KDM5B significantly improved the antitumor efficacy of antiangiogenic therapy in vivo. Taken together, PIAS4-mediated SUMOylation stabilized KDM5B protein by disturbing ubiquitination-dependent proteasomal degradation to overcome hypoxia stress. Targeting SUMOylation-dependent KDM5B upregulation might be considered when the antiangiogenic therapy was applied in cancer treatment.

12.
Am J Cancer Res ; 10(10): 3106-3126, 2020.
Article in English | MEDLINE | ID: mdl-33163260

ABSTRACT

Amino acid transporters mediate substrates across cellular membranes and their fine-tuned regulations are critical to cellular metabolism, growth, and death. As the functional component of system Xc-, which imports extracellular cystine with intracellular glutamate release at a ratio of 1:1, SLC7A11 has diverse functional roles in regulating many pathophysiological processes such as cellular redox homeostasis, ferroptosis, and drug resistance in cancer. Notably, accumulated evidence demonstrated that SLC7A11 is overexpressed in many types of cancers and is associated with patients' poor prognosis. As a result, SLC7A11 becomes a new potential target for cancer therapy. In this review, we first briefly introduce the structure and function of SLC7A11, then discuss its pathological role in cancer. We next summarize current available data of how SLC7A11 is subjected to fine regulations at multiple levels. We further describe the potential inhibitors of the SLC7A11 and their roles in human cancer cells. Finally, we propose novel insights for future perspectives on the modulation of SLC7A11, as well as possible targeted strategies for SLC7A11-based anti-cancer therapies.

13.
Cancer Manag Res ; 12: 10461-10468, 2020.
Article in English | MEDLINE | ID: mdl-33122945

ABSTRACT

BACKGROUND: This study was to investigate the prognostic factors of patients with advanced gastric cancer and described a sample model to better differentiate the patients who could better benefit from palliative chemotherapy. PATIENTS AND METHODS: In this retrospective study, 112 gastric cancer patients at stage IV following first-line chemotherapy were enrolled from July 2013 to September 2019. The clinical factors including age, sex, ECOG, pathologic types, metastatic sites, blood indexes, response of first-line chemotherapy, and survival were collected. The treatment responses were evaluated using the response evaluation criteria in solid tumors (RECIST). The survival curves were drawn by the Kaplan-Meier method, and the independent prognostic factors of overall survival (OS) were analyzed by Cox proportional hazards regression model. RESULTS: In this study, the median overall survival (mOS) of gastric cancer patients was 10.5 months, the disease remission rate (PR) was 21.4%, and the disease control rate (DCR) was 86.6%. Multivariate analysis identified 5 independent prognostic factors: peritoneal metastasis [P = 0.002; hazard risk (HR), 2.394; 95% CI 1.394-4.113], hemoglobin <90g/L [P = 0.001; hazard risk (HR), 2.674; 95% CI 1.536-4.655], LDH ≥225 U/L [P = 0.033; hazard risk (HR), 1.818; 95% CI 1.409-3.150], and 3 times higher level of CEA [P = 0.006; hazard risk (HR), 2.123; 95% CI 1.238-3.640] along with CA199 [P = 0.005; hazard risk (HR), 2.544; 95% CI 1.332-4.856] than upper limit of normal. Based on the obtained data, a prognostic index was constructed, dividing the patients into three risk groups: low (n = 67), intermediate (n = 35), and high-risk group (n = 10). The mOS for low, intermediate, and high-risk groups was 13.9 months (95% CI 10.7-17.1), 8.1 months (95% CI 5.7-10.4), and 3.9 months (95% CI 2.6-5.3), respectively, whereas the 1-year survival rate was 56.4%, 20.0%, and 0.0%, respectively (P < 0.001). CONCLUSION: This model should facilitate the prediction of treatment outcomes and then individualized treatment of advanced gastric cancer patients.

14.
Mol Cancer ; 19(1): 129, 2020 08 24.
Article in English | MEDLINE | ID: mdl-32838807

ABSTRACT

BACKGROUND: Heat shock transcription factor1 (HSF1) was overexpressed to promote glutaminolysis and activate mTOR in colorectal cancer (CRC). Here, we investigated the mechanism for cancer-specific overexpression of HSF1. METHODS: HSF1 expression was analyzed by chromatin immunoprecipitation, qRT-PCR, immunohistochemistry staining and immunoblotting. HSF1 translation was explored by polysome profiling and nascent protein analysis. Biotin pulldown and m6A RNA immunoprecipitation were applied to investigate RNA/RNA interaction and m6A modification. The relevance of HSF1 to CRC was analyzed in APCmin/+ and APCmin/+ HSF1+/-mice. RESULTS: HSF1 expression and activity were reduced after the inhibition of WNT/ß-catenin signaling by pyrvinium or ß-catenin knockdown, but elevated upon its activation by lithium chloride (LiCl) or ß-catenin overexpression. There are much less upregulated genes in HSF1-KO MEF treated with LiCl when compared with LiCl-treated WT MEF. HSF1 protein expression was positively correlated with ß-catenin expression in cell lines and primary tissues. After ß-catenin depletion, HSF1 mRNA translation was impaired, accompanied by the reduction of its m6A modification and the upregulation of miR455-3p, which can interact with 3'-UTR of HSF1 mRNA to repress its translation. Interestingly, inhibition of miR455-3p rescued ß-catenin depletion-induced reduction of HSF1 m6A modification and METTL3 interaction. Both the size and number of tumors were significantly reduced in APCmin/+ mice when HSF1 was genetically knocked-out or chemically inhibited. CONCLUSIONS: ß-catenin suppresses miR455-3p generation to stimulate m6A modification and subsequent translation of HSF1 mRNA. HSF1 is important for ß-catenin to promote CRC development. Targeting HSF1 could be a potential strategy for the intervention of ß-catenin-driven cancers.


Subject(s)
Adenosine/analogs & derivatives , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Heat Shock Transcription Factors/genetics , MicroRNAs/genetics , RNA, Messenger/genetics , beta Catenin/metabolism , Adenosine/metabolism , Animals , Apoptosis/genetics , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Disease Models, Animal , Humans , Methylation , Mice , Models, Biological , Protein Biosynthesis , RNA Interference , Xenograft Model Antitumor Assays
15.
Int J Biol Sci ; 16(7): 1194-1206, 2020.
Article in English | MEDLINE | ID: mdl-32174794

ABSTRACT

Metabolic reprogramming is a hallmark of cancer. Mammalian genome is characterized by pervasive transcription, generating abundant non-coding RNAs (ncRNAs). Long non-coding RNAs (lncRNAs) are freshly discovered functional ncRNAs exerting extensive regulatory impact through diverse mechanisms. Emerging studies have revealed widespread roles of lncRNAs in the regulation of various cellular activities, including metabolic pathways. In this review, we summarize the latest advances regarding the regulatory roles of lncRNAs in cancer metabolism, particularly their roles in mitochondrial function, glucose, glutamine, and lipid metabolism. Moreover, we discuss the clinical application and challenges of targeting lncRNAs in cancer metabolism. Understanding the complex and special behavior of lncRNAs will allow a better depiction of cancer metabolic networks and permit the development of lncRNA-based clinical therapies by targeting cancer metabolism.


Subject(s)
Neoplasms/metabolism , RNA, Long Noncoding/metabolism , Animals , Humans , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Neoplasms/genetics , RNA, Long Noncoding/genetics
16.
Mol Cell Oncol ; 6(5): e1618174, 2019.
Article in English | MEDLINE | ID: mdl-31528694

ABSTRACT

MLN4924, a small molecular inhibitor of NEDD8 (neuronal precursor cell-expressed developmentally downregulated protein 8) activating enzyme (NAE), blocks cullin neddylation to inactivate cullin-RING ligase. We found that MLN4924 has additional activities: it triggers EGFR dimerization and activation of RAS/MAPK and PI3K/AKT1 signals to stimulate tumor sphere formation and inhibit ciliogenesis; and it triggers PKM2 tetramerization to promote glycolysis.

17.
Theranostics ; 9(10): 2999-3013, 2019.
Article in English | MEDLINE | ID: mdl-31244938

ABSTRACT

Background & Aims: Dysregulation of metabolism plays an important role in the development and progression of cancers, while the underlying mechanisms remain largely unknown. This study aims to explore the regulation and relevance of glycolysis in chemoresistance of gastric cancer. Methods: Biochemical differences between chemoresistant and chemosensitive cancer cells were determined by metabolism profiling, microarray gene expression, PCR or western blotting. Cancer cell growth in vitro or in vivo were analyzed by viability, apoptosis and nude mice assay. Immunoprecipation was used to explore the interaction of proteins with other proteins or DNAs. Results: By metabolic and gene expression profiling, we found that pyruvate dehydrogenase kinase 3 (PDK3) was highly expressed to promote glycolysis in chemoresistant cancer cells. Its genetic or chemical inhibition reverted chemoresistance in vitro and in vivo. It was transcriptionally regulated by transcription factor HSF1 (Heat shock factor 1). Interestingly, PDK3 can localize in the nucleus and interact with HSF1 to disrupt its phosphorylation by GSK3ß. Since HSF1 was subjected to FBXW7-catalyzed polyubiquitination in a phosphorylation-dependent manner, PDK3 prevented HSF1 from proteasomal degradation. Thus, metabolic enzyme PDK3 and transcription factor HSF1 forms a positive feedback loop to promote glycolysis. As a result, inhibition of HSF1 impaired enhanced glycolysis and reverted chemoresistance both in vitro and in vivo. Conclusions: PDK3 forms a positive feedback loop with HSF1 to drive glycolysis in chemoresistance. Targeting this mitonuclear communication may represent a novel approach to overcome chemoresistance.


Subject(s)
Drug Resistance, Neoplasm , Heat Shock Transcription Factors/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Stomach Neoplasms/physiopathology , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Expression Profiling , Glycogen Synthase Kinase 3 beta/metabolism , Glycolysis , Humans , Metabolome , Mice, Nude , Phosphorylation , Protein Interaction Mapping , Protein Processing, Post-Translational , Proteolysis
18.
Cell Death Dis ; 10(5): 363, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31043584

ABSTRACT

Chemoresistance is one of the most important challenges in the clinical management of lung cancer. SIRT1 is a NAD dependent protein deacetylase and implicated in diverse cellular processes such as DNA damage repair, and cancer progression. SIRT1 is upregulated in chemoresistant lung cancer cells, genetic knockdown or chemical inhibition of SIRT1 reversed chemoresistance by enhancing DNA damage and apoptosis activation, accompanied with XRCC1 degradation. E3 ligase ß-TrCP catalyzed the poly-ubiquitination of XRCC1 to promote its proteasome-dependent degradation. SIRT1 bound and deacetylated XRCC1 at lysine K260, K298 and K431, preventing it from ß-TrCP-dependent ubiquitination. Mutations of these three lysine sites in XRCC1 abrogated the interaction with ß-TrCP and prolonged the half-life of XRCC1 protein. Here, we describes SIRT1 confers chemoresistance to lung cancer cells by deacetylating and stabilizing XRCC1. Therefore, targeting SIRT1 might be a new strategy to manage the chemoresistance of lung cancer, and probably other cancers.


Subject(s)
DNA, Neoplasm/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Sirtuin 1/genetics , X-ray Repair Cross Complementing Protein 1/genetics , beta-Transducin Repeat-Containing Proteins/genetics , Amino Acid Sequence , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Carbazoles/pharmacology , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Line, Tumor/pathology , Cisplatin/pharmacology , DNA Damage , DNA Repair/drug effects , DNA, Neoplasm/metabolism , Drug Resistance, Neoplasm/drug effects , Humans , Mice , Protein Processing, Post-Translational , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/metabolism , Ubiquitination , X-ray Repair Cross Complementing Protein 1/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism
19.
JCI Insight ; 4(4)2019 02 21.
Article in English | MEDLINE | ID: mdl-30668548

ABSTRACT

Abnormal activation of neddylation modification and dysregulated energy metabolism are frequently seen in many types of cancer cells. Whether and how neddylation modification affects cellular metabolism remains largely unknown. Here, we showed that MLN4924, a small-molecule inhibitor of neddylation modification, induces mitochondrial fission-to-fusion conversion in breast cancer cells via inhibiting ubiquitylation and degradation of fusion-promoting protein mitofusin 1 (MFN1) by SCFß-TrCP E3 ligase and blocking the mitochondrial translocation of fusion-inhibiting protein DRP1. Importantly, MLN4924-induced mitochondrial fusion is independent of cell cycle progression, but confers cellular survival. Mass-spectrometry-based metabolic profiling and mitochondrial functional assays reveal that MLN4924 inhibits the TCA cycle but promotes mitochondrial OXPHOS. MLN4924 also increases glycolysis by activating PKM2 via promoting its tetramerization. Biologically, MLN4924 coupled with the OXPHOS inhibitor metformin, or the glycolysis inhibitor shikonin, significantly inhibits cancer cell growth both in vitro and in vivo. Together, our study links neddylation modification and energy metabolism, and provides sound strategies for effective combined cancer therapies.


Subject(s)
Cyclopentanes/pharmacology , Energy Metabolism/drug effects , Mitochondria/drug effects , Neoplasms/drug therapy , Pyrimidines/pharmacology , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclopentanes/therapeutic use , Female , GTP Phosphohydrolases/metabolism , HEK293 Cells , Humans , Metformin/pharmacology , Mice , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Dynamics/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Naphthoquinones/pharmacology , Neoplasms/pathology , Oxidative Phosphorylation/drug effects , Proteolysis/drug effects , Pyrimidines/therapeutic use , Ubiquitin-Activating Enzymes/metabolism , Ubiquitination/drug effects , Xenograft Model Antitumor Assays
20.
Oncotarget ; 8(26): 42664-42672, 2017 Jun 27.
Article in English | MEDLINE | ID: mdl-28424408

ABSTRACT

Cancer cells reprogram metabolism to coordinate their rapid growth. They addict on glutamine metabolism for adenosine triphosphate generation and macromolecule biosynthesis. In this study, we report that glutamine deprivation retarded cell growth and induced prosurvival autophagy. Autophagy inhibition by chloroquine significantly enhanced glutamine starvation induced growth inhibition and apoptosis activation. Asparagine deprivation by L-asparaginase exacerbated growth inhibition induced by glutamine starvation and autophagy blockage. Similar to glutamine starvation, inhibition of glutamine metabolism with a chemical inhibitor currently under clinical evaluation was synthetically lethal with chloroquine and L-asparaginase, drugs approved for the treatment of malaria and leukemia, respectively. In conclusion, inhibiting glutaminolysis was synthetically lethal with autophagy inhibition and asparagine depletion. Therefore, targeting glutaminolysis could be a promising approach for colorectal cancer treatment.


Subject(s)
Asparagine/deficiency , Colonic Neoplasms/metabolism , Glutamine/metabolism , Asparagine/metabolism , Asparagine/pharmacology , Autophagy/drug effects , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , HEK293 Cells , Humans , Signal Transduction , Sulfides/pharmacology , Thiadiazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...