Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Biomed Pharmacother ; 174: 116486, 2024 May.
Article En | MEDLINE | ID: mdl-38520865

Recurrence and metastasis of gastric cancer is a major therapeutic challenge for treatment. The presence of cancer stem cells (CSCs) is a major obstacle to the success of current cancer therapy, often leading to treatment resistance and tumor recurrence and metastasis. Therefore, it is important to develop effective strategies to eradicate CSCs. In this study, we developed a combined therapeutic strategy of photothermal therapy (PTT) and gastric cancer stem cells (GCSCs) inhibition by successfully synthesizing nanoliposomes loaded with IR780 (photosensitizer) and EN4 (c-Myc inhibitor). The nanocomposites are biocompatible and exhibit superior photoacoustic (PA) imaging properties. Under laser irradiation, IR780-mediated PTT effectively and rapidly killed tumor cells, while EN4 synergistically inhibited the self-renewal and stemness of GCSCs by suppressing the expression and activity of the pluripotent transcription factor c-Myc, preventing the tumor progression of gastric cancer. This Nano-EN-IR@Lip is expected to be a novel clinical nanomedicine for the integration of gastric cancer diagnosis, treatment and prevention.


Liposomes , Neoplastic Stem Cells , Photosensitizing Agents , Photothermal Therapy , Stomach Neoplasms , Stomach Neoplasms/pathology , Stomach Neoplasms/therapy , Stomach Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Humans , Photothermal Therapy/methods , Animals , Cell Line, Tumor , Photosensitizing Agents/pharmacology , Photosensitizing Agents/administration & dosage , Indoles/pharmacology , Indoles/chemistry , Nanoparticles/chemistry , Mice, Nude , Combined Modality Therapy , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Nanocomposites/chemistry
2.
Biochim Biophys Acta Mol Basis Dis ; 1869(6): 166727, 2023 08.
Article En | MEDLINE | ID: mdl-37137433

Cancer-associated fibroblasts (CAFs) are the predominant stromal cells in the microenvironment and play important roles in tumor progression, including chemoresistance. However, the response of CAFs to chemotherapeutics and their effects on chemotherapeutic outcomes are largely unknown. In this study, we showed that epirubicin (EPI) treatment triggered ROS which initiated autophagy in CAFs, TCF12 inhibited autophagy flux and further promoted exosome secretion. Inhibition of EPI-induced reactive oxygen species (ROS) production with N-acetyl-L-cysteine (NAC) or suppression of autophagic initiation with short interfering RNA (siRNA) against ATG5 blunted exosome release from CAFs. Furthermore, exosome secreted from EPI-treated CAFs not only prevented ROS accumulation in CAFs but also upregulated the CXCR4 and c-Myc protein levels in recipient ER+ breast cancer cells, thus promoting EPI resistance of tumor cells. Together, the current study provides novel insights into the role of stressed CAFs in promoting tumor chemoresistance and reveal a new function of TCF12 in regulating autophagy impairment and exosome release.


Breast Neoplasms , Cancer-Associated Fibroblasts , Exosomes , Humans , Female , Cancer-Associated Fibroblasts/pathology , Breast Neoplasms/pathology , Epirubicin/pharmacology , Epirubicin/metabolism , Reactive Oxygen Species/metabolism , Drug Resistance, Neoplasm , Fibroblasts/metabolism , Exosomes/metabolism , Tumor Microenvironment , Basic Helix-Loop-Helix Transcription Factors/metabolism
3.
Cell Death Differ ; 28(9): 2708-2727, 2021 09.
Article En | MEDLINE | ID: mdl-33875796

The aberrant classical miRNAs are considered to play significant roles in tumor progression. However, it remains unclear for nonclassical miRNAs, a set of Drosha-independent miRNAs in the process of various biology. Here, we reveal that a nonclassical miR-4646-5p plays a pivotal role in gastric cancer (GC) metastasis. MiR-4646-5p, one of Drosha-independent mirtronic miRNA, is aberrant up-regulated in Drosha-low expressed GC and Drosha-knockdown gastric cancer cells. Mirtronic miR-4646-5p is a specific transcription splicing product of intron 3 of the host gene Abhd16a with the aid of SRSF2. The enhanced miR-4646-5p can stabilize HIF1A by targeting PHD3 to positive feedback regulate Abhd16a and miR-4646-5p itself expressions. ABHD16A, as an emerging phosphatidylserine-specific lipase, involves in lipid metabolism leading to lysophosphatidylserines (lyso-PSs) accumulation, which stimulates RhoA and downstream LIMK/cofilin cascade activity through GPR34/Gi subunit, thus causes metastasis of gastric cancer. In addition, miR-4646-5p/PHD3/HIF1A signaling can also up-regulate RhoA expression and synergistically promote gastric cancer cell invasion and metastasis. Our study provides new insights of nonclassical mirtronic miRNA on tumor progress and may serve as a new diagnostic biomarker for gastric cancer. MiR-4646-5p and its host gene Abhd16a mediated abnormal lipid metabolism may be a new target for clinical treatment of gastric cancer.


Lysophospholipids/metabolism , Metabolomics/methods , Monoacylglycerol Lipases/metabolism , Stomach Neoplasms/genetics , Animals , Humans , Male , Mice , Mice, Nude , Neoplasm Metastasis , Stomach Neoplasms/pathology , Transfection
4.
Cancer Lett ; 478: 8-21, 2020 05 28.
Article En | MEDLINE | ID: mdl-32142918

Drosha-dependent canonical microRNAs (miRNAs) play a crucial role in the biological functions and development of cancer. However, the effects of Drosha-independent non-canonical miRNAs remain poorly understood. In our previous work, we found a set of aberrant miRNAs, including some upregulated miRNAs, called Drosha-independent noncanonical miRNAs, in Drosha-knockdown gastric cancer (GC) cells. Surprisingly, Drosha-silenced GC cells still retained strong malignant properties (e.g., proliferation ability and cancer stem cell (CSC) characteristics), indicating that aberrantly upregulated non-canonical miRNAs may play an important role in the maintenance of the malignant properties in GC cells that express low Drosha levels. Here, we report that miR-6778-5p, a noncanonical miRNA, acts as a crucial regulator for maintenance of CSC stemness in Drosha-silenced GC cells. MiR-6778-5p belongs to the 5'-tail mirtron type of non-canonical miRNAs and is transcript splice-derived from intron 5 of SHMT1 (coding cytoplasmic serine hydroxymethyltransferase). It positively regulates expression of its host gene, SHMT1, via targeting YWHAE in Drosha-knockdown GC cells. Similar to its family member SHMT2, SHMT1 plays a crucial role in folate-dependent serine/glycine inter-conversion in one-carbon metabolism. In Drosha wild type GC cells, SHMT2 mediates a mitochondrial-carbon metabolic pathway, which is a major pathway of one-carbon metabolism in normal cells and most cancer cells. However, in Drosha-silenced or Drosha low-expressing GC cells, miR-6778-5p positively regulates SHMT1, instead of SHMT2, thus mediating a compensatory activation of cytoplasmic carbon metabolism that plays an essential role in the maintenance of CSCs in gastric cancer (GCSCs). Drosha wild type GCSCs with SHMT2 are sensitive to 5-fluorouracil; however, Drosha low-expressing GCSCs with SHMT1 are 5-FU-resistant. The loss of miR-6778-5p or SHMT1 notably mitigates GCSC sphere formation and increases sensitivity to 5-fluorouracil in Drosha-knockdown gastric cancer cells. Thus, our study reveals a novel function of Drosha-independent noncanonical miRNAs in maintaining the stemness of GCSCs.


Carbon/metabolism , Folic Acid/metabolism , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Ribonuclease III/genetics , Stomach Neoplasms/pathology , 14-3-3 Proteins/genetics , Animals , Cell Line, Tumor , Cytosol , Female , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glycine Hydroxymethyltransferase/genetics , Humans , Mice , Neoplasm Transplantation , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Up-Regulation
5.
Theranostics ; 7(16): 3972-3988, 2017.
Article En | MEDLINE | ID: mdl-29109792

Tumor microenvironment contributes to tumor angiogenesis. However, the role of the activated cancer associated-fibroblasts (CAFs) in angiogenesis is still unclear. Here we report that miR-205/YAP1 signaling in the activated stromal fibroblasts plays a critical role in VEGF-independent angiogenesis in breast tumor. Methods: miR-205 expression was assessed by quantitative real-time polymerase chain reaction (qRT-PCR); YAP1 expression by qRT-PCR, western blotting and immunohistochemistry; IL11 and IL15 expression by qRT-PCR, western blotting and ELISA. Tube formation and three-dimensioned sprouting assays in vitro, and orthotopic Xenografts in vivo were conducted as angiogenesis experiments. The mechanism of miR-205/YAP1-mediated tumor angiogenesis was analyzed via overexpression and shRNA, siRNA, or antibody neutralization experiments in combination with anti-VEGF antibody or Axitinib. Results: miR-205/YAP1 signaling axis activates breast normal fibroblasts (NFs) into CAFs, promotes tubule formation and sprouting of Human Umbilical Vein Endothelial Cells (HUVECs). Rescue of miR-205 in CAFs blunts angiogenesis processes. YAP1, a target of miR-205, does not regulate VEGF expression but specifically enhances IL11 and IL15 expressions, maintaining tumor angiogenesis even in the presence of Axitinib or after exhaustion of VEGF by neutralizing VEGF antibody. IL11 and IL15 released from CAFs activate STAT3 signaling in HUVECs. Blockage of IL11 and IL15 expression in CAFs results in the inactivation of STAT3-signaling in HUVECs and repression of the CAF-induced angiogenesis. The blunt angiogenesis halts the invasion and metastasis of breast cancer cells in vivo. Conclusions: These results provide a novel insight into breast CAF-induced tumor angiogenesis in a VEGF-independent manner.


Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , MicroRNAs/metabolism , Phosphoproteins/metabolism , STAT3 Transcription Factor/metabolism , Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/genetics , Female , Fibroblasts/metabolism , Humans , MicroRNAs/genetics , Neovascularization, Pathologic/metabolism , Phosphoproteins/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors , Vascular Endothelial Growth Factor A/metabolism , YAP-Signaling Proteins
6.
Cell Death Dis ; 8(3): e2642, 2017 03 02.
Article En | MEDLINE | ID: mdl-28252644

Drosha is an RNA III-like enzyme that has an aberrant expression in some tumors. Our previous studies showed the aberrant Drosha in gastric tumors. However, the roles of nuclear Drosha, the main regulator of microRNA (miRNA) biogenesis, in gastric cancer (GC) progression remain poorly understood. In this study, we demonstrated that nuclear Drosha is significantly associated with cell invasion of GC and that Drosha silence impedes the tumor invasion. Knockdown of Drosha led to a set of dysregulated miRNAs in GC cells. Multiple targets of these miRNAs were the members in cell migration, invasion and metastasis-associated signaling (e.g. ECM-receptor interaction, focal adhesion, p53 signaling and MAPK signaling pathway) revealed by bioinformatics analysis. LAMC2 (a key element of ECM-receptor signaling) and CD82 (a suppressor of p53 signaling) are the targets of miR-622 and miR-197, respectively. High levels of LAMC2 and low levels of CD82 were significantly related to the worse outcome for GC patients. Furthermore, overexpression of LAMC2 and knockdown of CD82 markedly promoted GC cell invasion and activated EGFR/ERK1/2-MMP7 signaling via upregulation of the expression of phosphorylated (p)-EGFR, p-ERK1/2 and MMP7. Our findings suggest that nuclear Drosha potentially has a role in the development of GC.


ErbB Receptors/metabolism , Kangai-1 Protein/metabolism , Laminin/metabolism , MAP Kinase Signaling System/physiology , Matrix Metalloproteinase 7/metabolism , MicroRNAs/metabolism , Ribonuclease III/metabolism , Stomach Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/physiology , Gene Expression Regulation, Neoplastic/physiology , Humans , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Phosphorylation/physiology , Signal Transduction/physiology , Stomach Neoplasms/pathology , Up-Regulation/physiology
7.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(9): 1207-11, 2016 Sep.
Article Zh | MEDLINE | ID: mdl-27609577

Objective To establish a gastric cancer cell line with stable Drosha silenced and explore the effect of Drosha on the chemosensitivity of gastric cancer cells to epirubicin. Methods Interfering sequences targeting Drosha were designed and inserted into the lentiviral vectors, which were used to transfect MGC-803 cells. The level of Drosha mRNA was detected by quantitative real-time PCR; Drosha protein was detected by Western blotting; MTT assay was performed to test the 50% inhibitory concentration (IC50) of epirubicin agaisnt wide-type MGC-803 cells. After the treatment with IC50 epirubicin, the apoptosis rate of each cell group was determined by flow cytometry; the expressions of apoptosis-related proteins caspase-3, caspase-9, Bax, Bcl-2 were assessed by Western blotting. Results The gastric cancer MGC-803 cells with stable Drosha silenced were successfully established, and the levels of Drosha mRNA and protein were reduced. After the cells were treated with 0.5 mg/L(IC50) epirubicin, the apoptosis rate of MGC-803 cells was raised, the protein expressions of caspase-3 , caspase-9 and Bax were significantly upregulated and Bcl-2 was downregulated. Conclusion The silence of Drosha expression can promote the sensitivity of gastric cancer to epirubicin.


Antineoplastic Agents/pharmacology , Epirubicin/pharmacology , Ribonuclease III/genetics , Stomach Neoplasms/genetics , Apoptosis/drug effects , Caspase 9/genetics , Caspase 9/metabolism , Cell Line, Tumor , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribonuclease III/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/physiopathology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
8.
Dig Dis Sci ; 61(4): 1080-90, 2016 Apr.
Article En | MEDLINE | ID: mdl-26694172

BACKGROUND: The nuclear localization of Drosha is critical for its function as a microRNA maturation regulator. Dephosphorylation of Drosha at serine 300 and serine 302 disrupts its nuclear localization, and aberrant distribution of Drosha has been detected in some tumors. AIMS: The purpose of the present study was to assess cytoplasmic/nuclear Drosha expression in gastric cancer carcinogenesis and progression. METHODS: Drosha expression and its subcellular location was investigated by immunohistochemical staining of a set of tissue microarrays composed of normal adjacent tissues (374), chronic gastritis (137), precancerous lesions (94), and gastric adenocarcinoma (829) samples, and in gastric cancer cell lines with varying differentiation by immunofluorescence and western blot assay. RESULTS: Gradual loss of cytoplasmic Drosha was accompanied by tumor progression in both gastric cancer tissues and cell lines, and was inversely associated with tumor volume (P = 0.002), tumor grade (P < 0.001), tumor stage (P = 0.018), and distant metastasis (P = 0.026). Aberrant high levels of cytoplasmic Drosha were apparent in intestinal metaplasia and dysplasia tissues. The levels of nuclear Drosha were sharply decreased in chronic gastritis and maintained through precancerous lesions to gastric cancer. High levels of cytoplasmic Drosha predicted longer survival (LR = 7.088, P = 0.008) in gastric cancer patients. CONCLUSIONS: Our data provide novel insights into gastric cancer that cytoplasmic Drosha potentially plays a role in preventing carcinogenesis and tumor progression, and may be an independent predictor of patient outcome.


Carcinoma/metabolism , Gastritis, Atrophic/metabolism , Precancerous Conditions/metabolism , Ribonuclease III/metabolism , Stomach Neoplasms/metabolism , Carcinoma/mortality , Carcinoma/pathology , Case-Control Studies , Cell Line, Tumor , China/epidemiology , Cytoplasm/metabolism , Disease Progression , Female , Gastric Mucosa/metabolism , Humans , Male , Metaplasia/metabolism , Middle Aged , Prognosis , Stomach/pathology , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Tissue Array Analysis
9.
Int J Biochem Cell Biol ; 71: 62-71, 2016 Feb.
Article En | MEDLINE | ID: mdl-26693891

Twist, a highly conserved basic Helix-Loop-Helix transcription factor, functions as a major regulator of epithelial-mesenchymal transition (EMT) and tumor metastasis. In different cell models, signaling pathways such as TGF-ß, MAPK/ERK, WNT, AKT, JAK/STAT, Notch, and P53 have also been shown to play key roles in the EMT process, yet little is known about the signaling pathways regulated by Twist in tumor cells. Using iTRAQ-labeling combined with 2D LC-MS/MS analysis, we identified 194 proteins with significant changes of expression in MCF10A-Twist cells. These proteins reportedly play roles in EMT, cell junction organization, cell adhesion, and cell migration and invasion. ECM-receptor interaction, MAPK, PI3K/AKT, P53 and WNT signaling were found to be aberrantly activated in MCF10A-Twist cells. Ingenuity Pathways Analysis showed that integrin ß1 (ITGB1) acts as a core regulator in linking integrin-linked kinase (ILK), Focal-adhesion kinase (FAK), MAPK/ERK, PI3K/AKT, and WNT signaling. Increased Twist and ITGB1 are associated with breast tumor progression. Twist transcriptionally regulates ITGB1 expression. Over-expression of ITGB1 or Twist in MCF10A led to EMT, activation of FAK/ILK, MAPK/ERK, PI3K/AKT, and WNT signaling. Knockdown of Twist or ITGB1 in BT549 and Hs578T cells decreased activity of FAK, ILK, and their downstream signaling, thus specifically impeding EMT and cell invasion. Knocking down ILK or inhibiting FAK, MAPK/ERK, or PI3K/AKT signaling also suppressed Twist-driven EMT and cell invasion. Thus, the Twist-ITGB1-FAK/ILK pathway and their downstream signaling network dictate the Twist-induced EMT process in human mammary epithelial cells and breast cancer cells.


Cell Movement , Epithelial-Mesenchymal Transition , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Integrin beta1/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Twist-Related Protein 1/metabolism , Cell Line, Tumor , Humans , Mammary Glands, Human/cytology , Neoplasm Invasiveness , Prognosis
10.
Stem Cells ; 34(1): 55-66, 2016 Jan.
Article En | MEDLINE | ID: mdl-26418365

Cancer stem cells (CSCs) are a subpopulation of neoplastic cells with self-renewal capacity and limitless proliferative potential as well as high invasion and migration capacity. These cells are commonly associated with epithelial-mesenchymal transition (EMT), which is also critical for tumor metastasis. Recent studies illustrate a direct link between EMT and stemness of cancer cells. Long non-coding RNAs (lncRNAs) have emerged as important new players in the regulation of multiple cellular processes in various diseases. To date, the role of lncRNAs in EMT-associated CSC stemness acquisition and maintenance remains unclear. In this study, we discovered that a set of lncRNAs were dysregulated in Twist-positive mammosphere cells using lncRNA microarray analysis. Multiple lncRNAs-associated canonical signaling pathways were identified via bioinformatics analysis. Especially, the Shh-GLI1 pathway associated lncRNA-Hh, transcriptionally regulated by Twist, directly targets GAS1 to stimulate the activation of hedgehog signaling (Hh). The activated Hh increases GLI1 expression, and enhances the expression of SOX2 and OCT4 to play a regulatory role in CSC maintenance. Thus, the mammosphere-formation efficiency (MFE) and the self-renewal capacity in vitro, and oncogenicity in vivo in Twist-positive breast cancer cells are elevated. lncRNA-Hh silence in Twist-positive breast cells attenuates the activated Shh-GLI1 signaling and decreases the CSC-associated SOX and OCT4 levels, thus reduces the MFE and tumorigenesis of transplanted tumor. Our results reveal that lncRNAs function as an important regulator endowing Twist-induced EMT cells to gain the CSC-like stemness properties.


Breast Neoplasms/metabolism , Hedgehog Proteins/metabolism , Neoplastic Stem Cells/metabolism , Nuclear Proteins/metabolism , RNA, Long Noncoding/metabolism , Signal Transduction , Twist-Related Protein 1/metabolism , Animals , Breast Neoplasms/pathology , Carcinogenesis/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/pathology , RNA, Long Noncoding/genetics , Spheroids, Cellular/pathology
11.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 31(11): 1519-22, 1527, 2015 Nov.
Article Zh | MEDLINE | ID: mdl-26522361

OBJECTIVE: To investigate the role of Drosha expression in the progression of gastric adenocarcinoma and its impact on the invasive ability of SGC-7901 human gastric cancer cells. METHODS: Drosha expression was detected in 889 gastric carcinoma samples on tissue microarrays by immunohistochemistry staining and quantified by Image-Pro Plus software. Statistical analysis was used to evaluate the correlations between Drosha expression and the clinicopathological characteristics of the 889 tumor cases or the outcomes of 309 gastric adenocarcinoma patients. Drosha was knocked down in SGC-7901 cells by small interfering RNA (siRNA), and cell invasive ability was assessed by Transwell(TM) assay. RESULTS: Drosha expression was the highest in the well differentiated gastric adenocarcinoma (median absorbance, 0.4195), and the lowest in the poorly differentiated samples. Drosha expression was significantly related to Laren classification, tumor size, tumor invasion depth, lymph node metastasis, tumor pathological grade and stage. Patients in Drosha positive group had a higher survival rate than those in Drosha negative group. Silencing Drosha in SGC-7901 cells resulted in an enhanced cell invasion. CONCLUSION: Drosha expression was reduced gradually with the degrading histological differentiation of gastric adenocarcinoma, and the knock-down of Drosha expression could promote gastric adenocarcinoma cell invasion.


Adenocarcinoma/pathology , Ribonuclease III/physiology , Stomach Neoplasms/pathology , Adenocarcinoma/chemistry , Adult , Aged , Humans , Middle Aged , Neoplasm Invasiveness , Ribonuclease III/analysis , Stomach Neoplasms/chemistry
12.
Oncotarget ; 6(28): 25755-69, 2015 Sep 22.
Article En | MEDLINE | ID: mdl-26342198

Twist, a key regulator of epithelial-mesenchymal transition (EMT), plays an important role in the development of a tumorigenic phenotype. Energy metabolism reprogramming (EMR), a newly discovered hallmark of cancer cells, potentiates cancer cell proliferation, survival, and invasion. Currently little is known about the effects of Twist on tumor EMR. In this study, we found that glucose consumption and lactate production were increased and mitochondrial mass was decreased in Twist-overexpressing MCF10A mammary epithelial cells compared with vector-expressing MCF10A cells. Moreover, these Twist-induced phenotypic changes were augmented by hypoxia. The expression of some glucose metabolism-related genes such as PKM2, LDHA, and G6PD was also found to be upregulated. Mechanistically, activated ß1-integrin/FAK/PI3K/AKT/mTOR and suppressed P53 signaling were responsible for the observed EMR. Knockdown of Twist reversed the effects of Twist on EMR in Twist-overexpressing MCF10A cells and Twist-positive breast cancer cells. Furthermore, blockage of the ß1-integrin/FAK/PI3K/AKT/mTOR pathway by siRNA or specific chemical inhibitors, or rescue of p53 activation can partially reverse the switch of glucose metabolism and inhibit the migration of Twist-overexpressing MCF10A cells and Twist-positive breast cancer cells. Thus, our data suggest that Twist promotes reprogramming of glucose metabolism in MCF10A-Twist cells and Twist-positive breast cancer cells via activation of the ß1-integrin/FAK/PI3K/AKT/mTOR pathway and inhibition of the p53 pathway. Our study provides new insight into EMR.


Breast Neoplasms/enzymology , Energy Metabolism , Glucose/metabolism , Nuclear Proteins/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Twist-Related Protein 1/metabolism , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Energy Metabolism/drug effects , Energy Metabolism/genetics , Epithelial-Mesenchymal Transition , Female , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation, Neoplastic , Humans , Integrin beta1/metabolism , Lactic Acid/metabolism , Mitochondria/enzymology , Mitochondria/pathology , Nuclear Proteins/genetics , Phenotype , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA Interference , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Time Factors , Transfection , Tumor Suppressor Protein p53/genetics , Twist-Related Protein 1/genetics
13.
Cell Cycle ; 14(12): 1908-24, 2015.
Article En | MEDLINE | ID: mdl-25970706

Abnormal proliferation is one characteristic of cancer-associated fibroblasts (CAFs), which play a key role in tumorigenesis and tumor progression. Oxidative stress (OS) is the root cause of CAFs abnormal proliferation. ATM (ataxia-telangiectasia mutated protein kinase), an important redox sensor, is involved in DNA damage response and cellular homeostasis. Whether and how oxidized ATM regulating CAFs proliferation remains unclear. In this study, we show that there is a high level of oxidized ATM in breast CAFs in the absence of double-strand breaks (DSBs) and that oxidized ATM plays a critical role in CAFs proliferation. The effect of oxidized ATM on CAFs proliferation is mediated by its regulation of cellular redox balance and the activity of the ERK, PI3K-AKT, and Wnt signaling pathways. Treating cells with antioxidant N-acetyl-cysteine (NAC) partially rescues the proliferation defect of the breast CAFs caused by ATM deficiency. Administrating cells with individual or a combination of specific inhibitors of the ERK, PI3K-AKT, and Wnt signaling pathways mimics the effect of ATM deficiency on breast CAF proliferation. This is mainly ascribed to the ß-catenin suppression and down-regulation of c-Myc, thus further leading to the decreased cyclinD1, cyclinE, and E2F1 expression and the enhanced p21(Cip1) level. Our results reveal an important role of oxidized ATM in the regulation of the abnormal proliferation of breast CAFs. Oxidized ATM could serve as a potential target for treating breast cancer.


Ataxia Telangiectasia Mutated Proteins/metabolism , Breast Neoplasms/metabolism , Fibroblasts/metabolism , Wnt Signaling Pathway , Antioxidants/chemistry , Cell Line, Tumor , Cell Proliferation , DNA Breaks, Double-Stranded , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Homeostasis , Humans , MAP Kinase Kinase 1/metabolism , Mitochondria/metabolism , Oxidation-Reduction , Oxidative Stress , Oxygen/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , beta Catenin/metabolism
...