Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
Add more filters

Publication year range
1.
Arch Pharm (Weinheim) ; 357(4): e2300526, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38294206

ABSTRACT

The phosphorylation of STAT3 plays a critical physiological role in the proliferation of rectal cancer. Hence, inhibiting STAT3 phosphorylation is an effective anticancer approach. In this work, we designed a novel 5-R'-1-naphthylmethylamide scaffold as a small molecule inhibitor of STAT3 phosphorylation. The results showed that 3D and 4D have exceptional inhibitory ability against three different colorectal cancer (CRC) cell lines, and can induce apoptosis of CRC cells by inhibiting STAT3 phosphorylation, while having no killing effect on normal human cells. 3D and 4D can inhibit STAT3 phosphorylation in a time- and concentration-dependent manner, and also inhibit the nuclear translocation of interleukin (IL)-6-induced STAT3. In the in vivo tumor model research, 4D significantly reduced the tumor volume of mice and had no drug toxicity on other organ tissues. Furthermore, molecular docking studies revealed that 3D and 4D had greater binding free energy when interacting with the STAT3 SH2 structural domain, and could establish H-π interaction modes. Dynamic simulation studies indicated that both compounds were able to bind tightly to STAT3.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Phosphorylation , Molecular Docking Simulation , Structure-Activity Relationship , STAT3 Transcription Factor/chemistry , STAT3 Transcription Factor/metabolism , Apoptosis , Cell Line, Tumor , Cell Proliferation , Antineoplastic Agents/chemistry
2.
Ren Fail ; 46(1): 2316259, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38345033

ABSTRACT

Acute kidney injury (AKI) can progress to renal fibrosis and chronic kidney disease (CKD), which reduces quality of life and increases the economic burden on patients. However, the molecular mechanisms underlying renal fibrosis following AKI remain unclear. This study tested the hypothesis that the Krüppel-like factor 4 (KLF4)/miR-101/Collagen alpha-1X (COL10A1) axis could inhibit epithelial-mesenchymal transition (EMT) and renal fibrosis after AKI in a mouse model of ischemia-reperfusion (I/R)-induced renal fibrosis and HK-2 cells by gene silencing, overexpression, immunofluorescence, immunohistochemistry, real-time quantitative PCR, Western blotting, dual-luciferase reporter assay, fluorescence in situ hybridization (FISH) and ELISA. Compared with the Sham group, I/R induced renal tubular and glomerular injury and fibrosis, and increased the levels of BUN, serum Scr and neutrophil gelatinase-associated lipocalin (NGAL), Col10a1 and Vimentin expression, but decreased E-cadherin expression in the kidney tissues of mice at 42 days post-I/R. Similarly, hypoxia promoted fibroblastic morphological changes in HK-2 cells and enhanced NGAL, COL10A1, Vimentin, and α-SMA expression, but reduced E-cadherin expression in HK-2 cells. These pathological changes were significantly mitigated in COL10A1-silenced renal tissues and HK-2 cells. KLF4 induces miR-101 transcription. More importantly, hypoxia upregulated Vimentin and COL10A1 expression, but decreased miR-101, KLF4, and E-cadherin expression in HK-2 cells. These hypoxic effects were significantly mitigated or abrogated by KLF4 over-expression in the HK-2 cells. Our data indicate that KLF4 up-regulates miR-101 expression, leading to the downregulation of COL10A1 expression, inhibition of EMT and renal fibrosis during the pathogenic process of I/R-related renal fibrosis.


Subject(s)
Acute Kidney Injury , MicroRNAs , Humans , Mice , Animals , MicroRNAs/metabolism , Lipocalin-2 , Vimentin/metabolism , Kruppel-Like Factor 4 , In Situ Hybridization, Fluorescence , Quality of Life , Cadherins/metabolism , Acute Kidney Injury/genetics , Epithelial-Mesenchymal Transition , Collagen/metabolism , Fibrosis , Hypoxia
3.
Respir Res ; 24(1): 76, 2023 Mar 13.
Article in English | MEDLINE | ID: mdl-36915129

ABSTRACT

PIWI-interacting RNA (piRNA) is a class of recently discovered small non-coding RNA molecules with a length of 18-33 nt that interacts with the PIWI protein to form the piRNA/PIWI complex. The PIWI family is a subfamily of Argonaute (AGO) proteins that also contain the AGO family which bind to microRNA (miRNA). Recently studies indicate that piRNAs are not specific to in the mammalian germline, they are also expressed in a tissue-specific manner in a variety of human tissues and participated in various of diseases, such as cardiovascular, neurological, and urinary tract diseases, and are especially prevalent in malignant tumors in these systems. However, the functions and abnormal expression of piRNAs in respiratory tract diseases and their underlying mechanisms remain incompletely understood. In this review, we discuss current studies summarizing the biogenetic processes, functions, and emerging roles of piRNAs in respiratory tract diseases, providing a reference value for future piRNA research.


Subject(s)
MicroRNAs , Neoplasms , Respiratory Tract Diseases , Animals , Humans , Piwi-Interacting RNA , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Neoplasms/metabolism , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Mammals/genetics , Mammals/metabolism
4.
Proc Natl Acad Sci U S A ; 117(8): 4347-4357, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32041891

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths worldwide and non-small cell lung cancer (NSCLC) accounts for over 80% of lung cancer cases. The RNA binding protein, QKI, belongs to the STAR family and plays tumor-suppressive functions in NSCLC. QKI-5 is a major isoform of QKIs and is predominantly expressed in NSCLC. However, the underlying mechanisms of QKI-5 in NSCLC progression remain unclear. We found that QKI-5 regulated microRNA (miRNA), miR-196b-5p, and its expression was significantly up-regulated in NSCLC tissues. Up-regulated miR-196b-5p promotes lung cancer cell migration, proliferation, and cell cycle through directly targeting the tumor suppressors, GATA6 and TSPAN12. Both GATA6 and TSPAN12 expressions were down-regulated in NSCLC patient tissue samples and were negatively correlated with miR-196b-5p expression. Mouse xenograft models demonstrated that miR-196b-5p functions as a potent onco-miRNA, whereas TSPAN12 functions as a tumor suppressor in NSCLC in vivo. QKI-5 bound to miR-196b-5p and influenced its stability, resulting in up-regulated miR-196b-5p expression in NSCLC. Further analysis showed that hypomethylation in the promoter region enhanced miR-196b-5p expression in NSCLC. Our findings indicate that QKI-5 may exhibit novel anticancer mechanisms by regulating miRNA in NSCLC, and targeting the QKI5∼miR-196b-5p∼GATA6/TSPAN12 pathway may enable effectively treating some NSCLCs.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , GATA6 Transcription Factor/genetics , Lung Neoplasms/metabolism , MicroRNAs/metabolism , Tetraspanins/genetics , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/physiopathology , Cell Line, Tumor , Cell Proliferation , Disease Progression , Down-Regulation , Female , GATA6 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/physiopathology , Mice , Mice, Nude , MicroRNAs/genetics , Tetraspanins/metabolism , Xenograft Model Antitumor Assays
5.
Biochem Biophys Res Commun ; 609: 31-38, 2022 06 18.
Article in English | MEDLINE | ID: mdl-35413537

ABSTRACT

Lung cancer is a part of the commonest malignancies with the highest mortality rate in cancer-related deaths worldwide. Signal transducer and activator of transcription 3 (STAT3) and cyclin-dependent kinases (CDKs) are promising prognostic marker and therapeutic target in cancers. Our previous study has demonstrated the closely relationship between CDK9 and STAT3 in lung cancer. The inhibition of cell viability and migration in vitro by AT7519 were evaluated using methyl thiazolyl tetrazolium (MTT) assay, clonogenic assay and scratch wound model. The cell cycle analysis was evaluated using flow cytometry analysis and western blotting analysis. The apoptotic-induced efficiency was assessed by flow cytometry analysis, hoechst 33342 staining, caspase-3 activity analysis and western blotting analysis. The roles of STAT3 in AT7519 treatment for lung cancer were assessed by docking model and western blotting analysis. The patient-derived xenograft (PDX) models were used to investigate the effect of AT7519 in vivo. In this study, we found that AT7519, a CDK inhibitor, reduced the viability of lung cancer cells in vitro and strongly suppressed tumor growth in PDX model. AT7519 blocked cell cycle progression and induced apoptosis by inhibiting IL-6/STAT3 pathway. Taken together, AT519 exhibits great anti-tumor effects in lung cancer, and the mechanism was related closely to IL-6/STAT3 signaling pathway, which suggests the important roles of STAT3 in CDKs inhibitors. AT7519 might be a novel potential therapeutic agent based on this rationale.


Subject(s)
Antineoplastic Agents , Lung Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Cell Proliferation , Humans , Interleukin-6/metabolism , Lung Neoplasms/metabolism , Piperidines , Pyrazoles , STAT3 Transcription Factor/metabolism , Signal Transduction
6.
J Transl Med ; 20(1): 525, 2022 11 12.
Article in English | MEDLINE | ID: mdl-36371217

ABSTRACT

BACKGROUND: Non-small cell lung cancer (NSCLC) is the most common lung cancer with high mortality across the world, but it is challenging to develop an effective therapy for NSCLC. Celastrol is a natural bioactive compound, which has been found to possess potential antitumor activity. However, the underlying molecular mechanisms of celastrol activity in NSCLC remain elusive. METHODS: Cellular function assays were performed to study the suppressive role of celastrol in human NSCLC cells (H460, PC-9, and H520) and human bronchial epithelial cells BEAS-2B. Cell apoptosis levels were analyzed by flow cytometry, Hoechst 33342, caspase-3 activity analysis, and western blot analysis. Intracellular reactive oxygen species (ROS) were analyzed by flow cytometry and fluorescence microscope. Expression levels of endoplasmic reticulum (ER) stress-related proteins and phosphorylated signal transducer and activator of transcription 3 (P-STAT3) were identified via western blot analysis. A heterograft model in nude mice was employed to evaluate the effect of celastrol in vivo. RESULTS: Celastrol suppressed the growth, proliferation, and metastasis of NSCLC cells. Celastrol significantly increased the level of intracellular ROS; thus, triggering the activation of the ER stress pathway and inhibition of the P-STAT3 pathway, and eventually leading to cell apoptosis, and the effects were reversed by the pre-treatment with N-Acetyl-L-cysteine (NAC). Celastrol also suppressed tumor growth in vivo. CONCLUSION: The outcomes revealed that celastrol plays a potent suppressive role in NSCLC in vitro and in vivo. Celastrol induces apoptosis via causing mitochondrial ROS accumulation to suppress the STAT3 pathway. Celastrol may have potential application prospects in the therapy of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Mice , Animals , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , STAT3 Transcription Factor/metabolism , Reactive Oxygen Species/metabolism , Mice, Nude , Lung Neoplasms/pathology , Apoptosis , Cell Line, Tumor , Cell Proliferation
7.
Pharmacol Res ; 183: 106389, 2022 09.
Article in English | MEDLINE | ID: mdl-35934193

ABSTRACT

Lung adenocarcinoma (LUAD) is associated with poor prognosis. Identifying novel cancer targets and helpful therapeutic strategies remains a serious clinical challenge. This study detected differentially expressed genes in The Cancer Genome Atlas (TCGA) LUAD data collection. We also identified a predictive DNA biomarker, G protein-coupled receptor 37 (GPR37), which was verified as a prognostic biomarker with a critical role in tumor progression. In human LUAD specimens and microarray analyses, we determined that GPR37 was significantly upregulated and associated with a poor prognosis. GPR37 downregulation markedly inhibited the proliferation and migration of LUAD both in vitro and in vivo. Mechanistically, GPR37 could bind to CDK6, thereby facilitating tumor progression in LUAD by inducing cell cycle arrest at the G1 phase. GPR37 also facilitates tumorigenesis in xenograft tumors in vivo. High-throughput screening for GPR37-targeted drugs was performed using the Natural Products Library, which revealed the potential of Hypocrellin B to inhibit GPR37 and cell growth in LUAD. We demonstrated that Hypocrellin B suppressed LUAD cell proliferation and migration both in vitro and in vivo via GPR37 inhibition. Collectively, our findings reveal the role of GPR37 in LUAD progression and migration and the potential of GPR37 as a target for the treatment of LUAD. Thus, the specific inhibition of GPR37 by the natural product Hypocrellin B may possess the potential for the treatment of LUAD.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Animals , Biomarkers , Cell Proliferation/physiology , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Precision Medicine , Prognosis , Receptors, G-Protein-Coupled
8.
J Nat Prod ; 85(4): 765-775, 2022 04 22.
Article in English | MEDLINE | ID: mdl-35200033

ABSTRACT

Non-small-cell lung carcer (NSCLC), the main histological subtype of lung cancer, is responsible for significant morbidity and mortality worldwide. Telocinobufagin, an active compound of the Chinese traditional medicine ChanSu, has antitumor effects, but its mechanism of action remains unknown. Therefore, we investigated the effect of telocinobufagin on NSCLC growth and metastasis and its possible mechanism of action, in vitro and in vivo. Cell proliferation, migration, and apoptosis were measured by methyl thiazol tetrazolium assay, colony formation, 5-ethynyl-2'-deoxyuridine incorporation, Transwell migration, wound healing, and flow cytometry analysis. A mouse xenograft model was used to evaluate tumor formation in vivo. Telocinobufagin was found to suppress proliferation and metastasis and induce apoptosis in human NSCLC cells. Moreover, telocinobufagin was able to significantly inhibit STAT3 phosphorylation at tyrosine 705 (Y705) and its downstream targets. Additionally, telocinobufagin also impaired the IL-6-induced nuclear translocation of STAT3. Consistent with the in vitro experiments, telocinobufagin reduced the A549 xenograft tumor burden and the levels of P-STAT3Y705, MCL1, BCL2, and cleaved PARP1 in vivo. These results support telocinobufagin as a promising STAT3 signaling inhibitor candidate for the treatment of NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Apoptosis , Bufanolides , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Humans , Lung Neoplasms/pathology , Mice , STAT3 Transcription Factor , Signal Transduction
9.
Acta Pharmacol Sin ; 43(7): 1633-1645, 2022 Jul.
Article in English | MEDLINE | ID: mdl-34811514

ABSTRACT

Cyclin-dependent kinase (CDK) 9 associates mainly with cyclin T1 and forms the positive transcription elongation factor b (p-TEFb) complex responsible for transcriptional regulation. It has been shown that CDK9 modulates the expression and activity of oncogenes, such as MYC and murine double minute 4 (MDM4), and it also plays an important role in development and/or maintenance of the malignant cell phenotype. Malfunction of CDK9 is frequently observed in numerous cancers. Recent studies have highlighted the function of CDK9 through a variety of mechanisms in cancers, including the formation of new complexes and epigenetic alterations. Due to the importance of CDK9 activation in cancer cells, CDK9 inhibitors have emerged as promising candidates for cancer therapy. Natural product-derived and chemically synthesized CDK9 inhibitors are being examined in preclinical and clinical research. In this review, we summarize the current knowledge on the role of CDK9 in transcriptional regulation, epigenetic regulation, and different cellular factor interactions, focusing on new advances. We show the importance of CDK9 in mediating tumorigenesis and tumor progression. Then, we provide an overview of some CDK9 inhibitors supported by multiple oncologic preclinical and clinical investigations. Finally, we discuss the perspective and challenge of CDK9 modulation in cancer.


Subject(s)
Cyclin-Dependent Kinase 9 , Neoplasms , Animals , Cyclin T/genetics , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Epigenesis, Genetic , Gene Expression Regulation , Mice , Neoplasms/metabolism , Positive Transcriptional Elongation Factor B/metabolism , Transcription, Genetic
10.
J Cell Mol Med ; 25(2): 801-812, 2021 01.
Article in English | MEDLINE | ID: mdl-33259114

ABSTRACT

Colorectal cancer (CRC) accounts for about 10% of all annually diagnosed cancers and cancer-related deaths worldwide. STAT3 plays a vital role in the occurrence and development of tumours. Gracillin has shown a significant antitumour activity in tumours, but its mechanism remains unknown. The human CRC cell lines HCT116, RKO, and SW480 and immunodeficient mice were used as models to study the effects of gracillin on cell proliferation, migration and apoptosis. These were evaluated by cell viability, colony formation, wound-healing migration and cell apoptosis assays. Luciferase reporter assay, and immunostaining and western blot analyses were used to explore the specific mechanism through which gracillin exerts its effects. Gracillin significantly reduces viability and migration and stimulates apoptosis in human CRC cells. It also significantly inhibits tumour growth with no apparent physiological toxicity in animal model experiments. Moreover, gracillin is found to inhibit STAT3 phosphorylation and STAT3 target gene products. In addition, gracillin inhibits IL6-induced nuclear translocation of P-STAT3. Gracillin shows potent efficacy against CRC by inhibiting the STAT3 pathway. It should be further explored as a unique STAT3 inhibitor for the treatment of CRC.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Spirostans/pharmacology , Spirostans/therapeutic use , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , HCT116 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Wound Healing/drug effects , Xenograft Model Antitumor Assays
11.
Br J Cancer ; 124(3): 645-657, 2021 02.
Article in English | MEDLINE | ID: mdl-33122847

ABSTRACT

BACKGROUND: Cyclin-dependent kinase 9 (CDK9) is a promising prognostic marker and therapeutic target in cancers. Bufalin is an effective anti-tumour agent; however, the clinical application of bufalin is limited due to its high toxicity. Acetyl-bufalin, the bufalin prodrug, was designed and synthesised with higher efficiency and lower toxicity. METHODS: Three non-small-cell lung cancer (NSCLC) cell lines, a xenograft model and a patient-derived xenograft (PDX) model were used to examine the effects of acetyl-bufalin. CDK9/STAT3 involvement was investigated by knockdown with siRNA, proteome microarray assay, western blot analysis and co-immunoprecipitation experiments. Acute toxicity test and pharmacokinetics (PK) study were conducted to assess the safety and PK. The human NSCLC tissues were analysed to verify high CDK9 expression. RESULTS: We showed that CDK9 induced NSCLC cell proliferation and that this effect was associated with STAT3 activation, specifically an increase in STAT3 phosphorylation and transcription factor activity. Acetyl-bufalin is an effective and safety inhibitor of the CDK9/STAT3 pathway, leading to the impediment of various oncogenic processes in NSCLC. Molecular docking and high-throughput proteomics platform analysis uncovered acetyl-bufalin directly binds to CDK9. Consequently, acetyl-bufalin impaired the complex formation of CDK9 and STAT3, decreased the expressions of P-STAT3, and transcribed target genes such as cyclin B1, CDC2, MCL-1, Survivin, VEGF, BCL2, and it upregulated the expression levels of BAX and caspase-3 activity. Acetyl-bufalin inhibited tumour growth in NSCLC xenograft and PDX models. CONCLUSIONS: Acetyl-bufalin is a novel blocker of the CDK9/STAT3 pathway thus may have potential in therapy of NSCLC and other cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Bufanolides/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Lung Neoplasms/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Bufanolides/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Enzyme Inhibitors/pharmacology , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Heterografts , Humans , Lung Neoplasms/metabolism , M Phase Cell Cycle Checkpoints/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Molecular Docking Simulation , Prodrugs/pharmacology , RNA, Small Interfering/genetics , Rats , STAT3 Transcription Factor/metabolism
12.
BMC Nephrol ; 22(1): 285, 2021 08 23.
Article in English | MEDLINE | ID: mdl-34425760

ABSTRACT

BACKGROUND: Crescentic glomerulonephritis is a disease characterized by severe glomerular injuries that is classified into five different pathological types. Patients with type V disease have pauci-immune crescentic glomerulonephritis (PICGN) that is negative for anti-neutrophil cytoplasmic autoantibodies (ANCAs). There are limited clinical data on the manifestations, treatment, and prognosis of type V crescentic glomerulonephritis, especially in children. CASE PRESENTATION: A 13-year-old girl who had an intermittent fever for more than 10 months was admitted to our hospital. She had no gross hematuria, oliguria, edema, or hypertension, but further tests indicated a decreased glomerular filtration rate, hematuria, proteinuria, and an elevated level of IL-6. The antinuclear antibody spectrum test was positive at 1:1000, and the ANCA and anti-glomerular basement membrane antibody tests were negative. A renal biopsy confirmed the diagnosis of ANCA-negative PICGN. We administered methylprednisolone pulse therapy with intravenous cyclophosphamide and oral mycophenolate mofetil. At the 3-month follow-up, her urine protein level was significantly lower, and her serum creatinine level was in the normal range. CONCLUSIONS: Fever may be an extrarenal manifestation of ANCA-negative PICGN, and IL-6 may play a role in the pathogenesis of this disease. Early methylprednisolone pulse therapy with an immunosuppressant may reduce symptoms and improve prognosis.


Subject(s)
Antibodies, Antineutrophil Cytoplasmic/blood , Glomerulonephritis/immunology , Interleukin-6/blood , Kidney Glomerulus/pathology , Adolescent , Female , Fever of Unknown Origin/etiology , Glomerulonephritis/complications , Glomerulonephritis/pathology , Humans , Kidney Glomerulus/ultrastructure
13.
J Cell Mol Med ; 24(2): 1568-1577, 2020 01.
Article in English | MEDLINE | ID: mdl-31778025

ABSTRACT

Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) is a bifunctional enzyme located in the mitochondria. It has been reported to be overexpressed in several malignancies. However, the relationship between the expression of MTHFD2 and non-small cell lung cancer (NSCLC) remains largely unknown. In this study, we found that MTHFD2 was significantly overexpressed in NSCLC tissues and cell lines. Knockdown of MTHFD2 resulted in reduced cell growth and tumorigenicity in vitro and in vivo. Besides, the mRNA and protein expression level of cell cycle genes, such as CCNA2, MCM7 and SKP2, was decreased in MTHFD2 knockdown H1299 cells. Our results indicate that the inhibitory effect of MTHFD2 knockdown on NSCLC may be mediated via suppressing cell cycle-related genes. These findings delineate the role of MTHFD2 in the development of NSCLC and may have potential applications in the treatment of NSCLC.


Subject(s)
Aminohydrolases/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/genetics , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Multifunctional Enzymes/genetics , Aminohydrolases/metabolism , Animals , Apoptosis/genetics , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Disease Progression , Female , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mice, Inbred BALB C , Mice, Nude , Multifunctional Enzymes/metabolism , Oncogenes , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation/genetics
14.
Am J Physiol Renal Physiol ; 319(6): F1125-F1134, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33135476

ABSTRACT

Renal interstitial fibrosis (RIF) is characterized by excessive extracellular matrix deposition and involves epithelial-mesenchymal transition (EMT). The lncRNA taurine-upregulated gene 1 (TUG1) participates in EMT in several cancers; however, the effect and underlying mechanism of TUG1 in RIF-related EMT remain unclear. Here, we explored the mechanisms by which TUG1 modulates RIF. An in vivo model of renal fibrosis was established by unilateral ureteral obstruction in Balb/c mice. Human renal proximal tubular epithelial (HK-2) cells treated with transforming growth factor (TGF)-ß1 were used to induce the in vitro model. Morphological changes and TUG1 expression were assessed. HK-2 cells were transfected with siRNA to silence TUG1. Western blot analysis, immunofluorescence staining, cell proliferation, and migration assays were performed to examine TGF-ß1-induced changes in EMT markers and EMT-like cell behaviors. TUG1 and ß-catenin (CTNNB1) levels were significantly upregulated, whereas miR-141-3p was significantly downregulated, during EMT in vitro and in vivo. TUG1 knockdown or miR-141-3p overexpression supported the epithelioid morphology of HK-2 cells while enhancing the downregulation of E-cadherin and upregulation of vimentin, α-smooth muscle actin, and ß-catenin levels in TGF-ß1-treated HK-2 cells. TUG1 knockdown promoted the proliferation and decreased the migration of HK-2 cells and enhanced the downregulation of miR-141-3p levels in TGF-ß1-treated HK-2 cells. TUG1 directly targeted miR-141-3p, and miR-141-3p was directly bound to CTNNB1. Downregulation of miR-141-3p inhibited TUG1 silencing-induced suppression of EMT. In conclusion, TUG1 promotes EMT in TGF-ß1-induced HK-2 cells via upregulation of ß-catenin levels by sponging miR-141-3p, suggesting a novel therapeutic candidate for RIF.


Subject(s)
Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Kidney Diseases/metabolism , Kidney Tubules/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , beta Catenin/metabolism , Animals , Cell Line , Cell Movement , Cell Proliferation , Disease Models, Animal , Epithelial Cells/pathology , Fibrosis , Humans , Kidney Diseases/etiology , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Tubules/pathology , Mice, Inbred BALB C , MicroRNAs/genetics , RNA Interference , RNA, Long Noncoding/genetics , Signal Transduction , Ureteral Obstruction/complications , beta Catenin/genetics
15.
J Cell Mol Med ; 23(3): 2194-2206, 2019 03.
Article in English | MEDLINE | ID: mdl-30609207

ABSTRACT

Triple-negative breast cancer (TNBC) is a subtype of breast cancer with poor clinical outcome and currently no effective targeted therapies are available. Alantolactone (ATL), a sesquiterpene lactone, has been shown to have potential anti-tumour activity against various cancer cells. However, the underlying mechanism and therapeutic effect of ATL in the TNBC are largely unknown. In the present study, we found that ATL suppresses TNBC cell viability by reactive oxygen species (ROS) accumulation and subsequent ROS-dependent endoplasmic reticulum (ER) stress both in vitro and in vivo. Thioredoxin reductase 1 (TrxR1) expression and activity of were significantly up-regulated in the TNBC tissue specimens compare to the normal adjacent tissues. Further analyses showed that ATL inhibits the activity of TrxR1 both in vitro and in vivo in TNBC and knockdown of TrxR1 in TNBC cells sensitized ATL-induced cell apoptosis and ROS increase. These results will provide pre-clinical evidences that ATL could be a potential therapeutic agent against TNBC by promoting ROS-ER stress-mediated apoptosis through partly targeting TrxR1.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Lactones/pharmacology , Sesquiterpenes, Eudesmane/pharmacology , Thioredoxin Reductase 1/genetics , Thioredoxin-Disulfide Reductase/genetics , Triple Negative Breast Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Endoplasmic Reticulum Stress/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred BALB C , Mice, Nude , Reactive Oxygen Species/metabolism , Thioredoxin Reductase 1/metabolism , Thioredoxin-Disulfide Reductase/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
16.
Mol Carcinog ; 58(11): 2026-2039, 2019 11.
Article in English | MEDLINE | ID: mdl-31397502

ABSTRACT

Carboxypeptidase A4 (CPA4) is a member of the metallocarboxypeptidase family. A previous study indicated that CPA4 may participate in the modulation of peptide hormone activity and hormone-regulated tissue growth and differentiation. However, the role of CPA4 in lung tumorigenesis remains unclear. Our study revealed that CPA4 expression was higher in both lung cancer cells and tumor tissues. We performed 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assays, colony-formation assays, and Cellomics ArrayScan Infinity analysis to demonstrate that CPA4 knockdown inhibited non small-cell lung cancer (NSCLC) cell proliferation. Conversely, ectopic expression of CPA4 enhanced lung cancer cell proliferation. Consistent with these observations, we generated xenograft tumor models to confirm that CPA4 downregulation suppressed NSCLC cell growth. Mechanistically, we revealed that CPA4 downregulation may induce apoptosis and G1-S arrest by suppressing the protein kinase B/c-MYC pathway. These results suggest that CPA4 has an oncogenic effect on lung cancer growth. Taken together, we identified a novel gene in lung cancer that might provide a basis for new therapeutic targets.


Subject(s)
Carboxypeptidases A/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Oncogene Protein v-akt/genetics , Proto-Oncogene Proteins c-myc/genetics , Animals , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Mice , Signal Transduction/genetics
17.
Mol Carcinog ; 58(4): 565-576, 2019 04.
Article in English | MEDLINE | ID: mdl-30520143

ABSTRACT

Several studies have implicated the feedback activation of signal transducer and activator of transcription 3 (STAT3) as a new cancer drug-resistance mechanism and linked it to the failure of epidermal growth factor receptor (EGFR)-targeted therapies. In this study, we discovered that Alantolactone, a natural sesquiterpene lactone, potently inhibited human pancreatic cancer cells and suppressed constitutively activated STAT3. In contrast, Alantolactone had little effect on the EGFR pathway. Moreover, combination of Alantolactone and an EGFR inhibitor, Erlotinib or Afatinib, demonstrated a remarkable synergistic anti-cancer effect against pancreatic cancer cells both in vitro and in vivo. Our results suggested that Alantolactone could sensitize human pancreatic cancer cells to EGFR inhibitors possibly through down-regulating the STAT3 signaling. Alantolactone, when combined with other EGFR targeted agents, could be further developed as a potential therapy for pancreatic cancer.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Lactones/pharmacology , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Sesquiterpenes, Eudesmane/pharmacology , Animals , Apoptosis , Cell Cycle , Cell Movement , Cell Proliferation , ErbB Receptors/antagonists & inhibitors , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , STAT3 Transcription Factor , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Mol Carcinog ; 58(8): 1512-1525, 2019 08.
Article in English | MEDLINE | ID: mdl-31069881

ABSTRACT

Lung cancer is a leading cause of cancer-related death worldwide. Cyanopyridines and aminocyanopyridines with carbon-nitrogen bonds have been proved to exert significant anticancer, antibacterial, and anti-inflammatory effects. In this study, we showed that aminocyanopyridine 3o and 3k displaying potent antitumor activity via inhibiting the signal transducer and activator of transcription 3 (STAT3) pathway. They blocked the constitutive STAT3 phosphorylation in a dose- and time-dependent manner and regulated the transcription of STAT3 target genes encoding apoptosis factors. Most importantly, 3o also inhibited interleukin-6-induced STAT3 activation and nuclear localization. Furthermore, 3o significantly inhibited the tumor growth of H460-derived xenografts. Taken together, these findings suggest that 3o and 3k are promising therapeutic drug candidates for lung cancer by inhibiting persistent STAT3 signaling.


Subject(s)
Antineoplastic Agents/pharmacology , Lung Neoplasms/drug therapy , Pyridines/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , A549 Cells , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Interleukin-6/metabolism , Janus Kinase 2/biosynthesis , Janus Kinase 3/biosynthesis , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Docking Simulation , Phosphorylation/drug effects , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
19.
Nephrology (Carlton) ; 24(2): 263-271, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29280536

ABSTRACT

AIM: The aim of the present study was to screen and verify downstream genes involved in the epithelial mesenchymal transition (EMT) induced by paired box 2 (PAX2) in NRK-52E cells. METHODS: NRK-52E cells were transfected with lentivirus carrying PAX2 gene or no-load virus respectively. Total RNA was isolated 72 h after transfection from PAX2-overexpressing cells and control cells. Isolated RNA was then hybridized with the Rat OneArray Plus expression profile chip. The chips were examined by Agilent 0.1 XDR to screen for differentially expressed genes, which were further analyzed to investigate complement-related genes as genes of interest. RESULTS: In NRK-52E cells, PAX2 overexpression promoted EMT followed by upregulation of 298 genes and downregulation of 293 genes. KEGG analysis indicated the differential expression of genes related to cytokines and their receptors, extracellular matrix (ECM), MAPKs, local adhesion, cancer, the complement cascade, and coagulation. Gene oncology analysis screened out genes related to molecular functions (e.g., hydrolase activity, phospholipase activity, components of the ECM) and biological processes (e.g., cell development, signal transduction, phylogeny), and cell components (e.g., cytoplasm, cell membrane, and ECM). Analysis of the complement system revealed upregulation of C3 and downregulation of CD55 and complement regulator factor H (CFH). CONCLUSION: PAX2 overexpression upregulates EMT in vitro and may regulate C3, CD55, and CFH.


Subject(s)
Complement System Proteins/metabolism , Epithelial-Mesenchymal Transition , Kidney Tubules/metabolism , PAX2 Transcription Factor/metabolism , Animals , Blotting, Western , CD55 Antigens/genetics , CD55 Antigens/metabolism , Cell Line , Complement C3/genetics , Complement C3/metabolism , Complement Factor H/genetics , Complement Factor H/metabolism , Complement System Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling/methods , Gene Expression Regulation , Kidney Tubules/pathology , Oligonucleotide Array Sequence Analysis , PAX2 Transcription Factor/genetics , Rats , Real-Time Polymerase Chain Reaction , Signal Transduction
20.
Anticancer Drugs ; 28(4): 392-400, 2017 04.
Article in English | MEDLINE | ID: mdl-28067673

ABSTRACT

Pancreatic cancer is the most commonly diagnosed malignancy among solid tumors and has shown an increasing trend year by year. Thus, there is an urgent need for the discovery of new anticancer drugs for the treatment of pancreatic cancer. In recent years, it has been reported that the compound HO-3867, a novel analog of the natural product curcumin, showed antitumor activity with low toxicity. However, the underlying mechanism of this compound's attack on cancer cells is not very clear. In the present study, it was found that HO-3867 showed good antitumor activity at the concentration of 2 µmol/l in PANC-1 and BXPC-3 cells. Importantly, it was also found that HO-3867 treatment significantly induced reactive oxygen species (ROS) production in human pancreatic cancer cell lines, inducing PANC-1 and BXPC-3 cells. Co-treatment with the ROS scavenger, N-acetyl cysteine, partially abrogated HO-3867-induced cell apoptosis. The activation of mitogen-activated protein kinase and endoplasmic reticulum stress indicated a downstream event of ROS generation in mediating the anticancer effect of the HO-3867. In addition, independent of the ROS pathway, direct STAT3 inhibition was observed in HO-3867-induced cell apoptosis. Taken together, the results of this work suggest that both the ROS-dependent ER stress and STAT3 pathways were implicated in the cell apoptosis induced by the novel compound HO-3867.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Pancreatic Neoplasms/drug therapy , Piperidones/pharmacology , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Apoptosis/drug effects , Cell Line, Tumor , Humans , MAP Kinase Signaling System/drug effects , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , STAT3 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL