Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Redox Biol ; 68: 102952, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37944384

ABSTRACT

Tamoxifen (TAM) resistance remains a major obstacle in the treatment of advanced breast cancer (BCa). In addition to the competitive inhibition of the estrogen receptor (ER) signaling pathway, damping of mitochondrial function by increasing reactive oxygen species (ROS) is critical for enhancing TAM pharmacodynamics. Here, we showed that RelB contributes to TAM resistance by inhibiting TAM-provoked ferroptosis. TAM-induced ROS level promoted ferroptosis in TAM-sensitive cells, but the effect was alleviated in TAM-resistant cells with high constitutive levels of RelB. Mechanistically, RelB inhibited ferroptosis by transcriptional upregulating glutathione peroxidase 4 (GPX4). Consequently, elevating RelB and GPX4 in sensitive cells increased TAM resistance, and conversely, depriving RelB and GPX4 in resistant cells decreased TAM resistance. Furthermore, suppression of RelB transcriptional activation resensitized TAM-resistant cells by enhancing ferroptosis in vitro and in vivo. The inactivation of GPX4 in TAM-resistant cells consistently resensitized TAM by increasing ferroptosis-mediated cell death. Together, this study uncovered that inhibition of ferroptosis contributes to TAM resistance of BCa via RelB-upregulated GPX4.


Subject(s)
Breast Neoplasms , Ferroptosis , Humans , Female , Tamoxifen/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Cell Death
2.
Free Radic Biol Med ; 164: 85-95, 2021 02 20.
Article in English | MEDLINE | ID: mdl-33418113

ABSTRACT

Adriamycin (ADR) resistance poses a significant challenge for successfully treating breast cancer (BCa). The mechanism underlying intrinsically acquisition of the resistance remains to be fully elucidated. Here, we describe that small extracellular vesicles (sEVs) mediated Hsp70 transfer is implicated in ADR resistance. The resistant cells derived sEVs were incubated with sensitive cells, thereby transmitting the resistant phenotype to the recipient cells. The internalization of the sEVs in the recipient cells and sEV-mediated Hsp70 transfer into mitochondria were examined by confocal microscope and transmission electron microscopy (TEM). Oxygen consumption rate (OCR) incorporated with extracellular acidification rate (ECAR) was quantified by Seahorse XF Analyzer. Mechanistically, sEVs transported Hsp70, leading to increased reactive oxygen species (ROS) and impaired mitochondria in the recipient cells, thereby inhibiting respiration but promoting glycolysis. The sEVs effect on the metabolism of the recipient cells was alleviated by silencing Hsp70 in sEVs donor cells. The aspect of sEV-Hsp70 on drug-resistant transmission was further validated by tumor zebrafish xenografts. The finding from this work suggests that sEV-mediated Hsp70 intercellular delivery enhances ADR resistance mainly through reprogramming the recipient cell energy metabolism.


Subject(s)
Breast Neoplasms , Extracellular Vesicles , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Doxorubicin/metabolism , Doxorubicin/pharmacology , Extracellular Vesicles/metabolism , Female , Humans , Mitochondria , Zebrafish
3.
Mol Oncol ; 15(5): 1528-1542, 2021 05.
Article in English | MEDLINE | ID: mdl-33508878

ABSTRACT

Chemotherapeutic resistance is a major obstacle in the control of advanced breast cancer (BCa). We have previously shown that small extracellular vesicles (sEVs) can transmit adriamycin resistance between BCa cells. Here, we describe that sEV-mediated TGF-ß1 intercellular transfer is involved in the drug-resistant transmission. sEVs were isolated and characterized from both sensitive and resistant cells. sEVs derived from the resistant cells were incubated with the sensitive cells and resulted in transmitting the resistant phenotype to the recipient cells. Cytokine antibody microarray revealed that most metastasis-associated cytokines present at the high levels in sEVs from the resistant cells compared with their levels in sEVs from the sensitive cells, particularly TGF-ß1 is enriched in sEVs from the resistant cells. The sEV-mediated TGF-ß1 intercellular transfer led to increasing Smad2 phosphorylation and improving cell survival by suppressing apoptosis and enhancing cell mobility. Furthermore, sEV-mediated drug-resistant transmission by delivering TGF-ß1 was validated using a zebrafish xenograft tumor model. These results elaborated that sEV-mediated TGF-ß1 intercellular transfer contributes to adriamycin resistance in BCa.


Subject(s)
Breast Neoplasms/drug therapy , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm , Extracellular Vesicles/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Antibiotics, Antineoplastic/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Embryo, Nonmammalian , Extracellular Vesicles/physiology , Female , Humans , MCF-7 Cells , Paracrine Communication/genetics , Transforming Growth Factor beta1/genetics , Tumor Microenvironment/genetics , Zebrafish/embryology
4.
Mol Ther Nucleic Acids ; 13: 64-77, 2018 Dec 07.
Article in English | MEDLINE | ID: mdl-30240971

ABSTRACT

Radioresistance remains to be a major obstacle in the management of patients with advanced prostate cancer (PCa). We have identified a mature miR-17-3p processed from the 3' arm of precursor miR-17, which appeared to be able to inhibit three major antioxidant enzymes located in mitochondria, i.e., manganese superoxide dismutase (MnSOD), glutathione peroxidase 2 (Gpx2), and thioredoxin reductase 2 (TrxR2). Here we show that upregulation of miR-17-3p remarkably sensitized PCa cells to ionizing radiation (IR). Reductions of the three antioxidants led to increasing cellular reactive oxygen species (ROS) accumulation as well as declining mitochondrial respiration. The miR-17-3p-mediated dysfunction of mitochondrial antioxidants apparently sensitizing IR therapy was manifested in vitro and in vivo. Substantially, the miR-17-3p effect on suppression of the antioxidants can be efficiently eliminated or attenuated by transfecting with either an miR-17-3p inhibitor or each of the related antioxidant cDNA expression constructs. Overall, in addition to the insights into the functional assessments for the duplex of miR-17-5p and miR-17-3p, the present study highlights the rigorous evidence that demonstrated suppression of multiple mitochondrial antioxidants by a single microRNA (miRNA), thereby providing a promising approach to improve radiotherapy for advanced PCa by targeting mitochondrial function.

5.
J Exp Clin Cancer Res ; 37(1): 174, 2018 Jul 27.
Article in English | MEDLINE | ID: mdl-30053873

ABSTRACT

BACKGROUND: The development of radioresistance is one of main causes for therapeutic failure of prostate cancer (PCa). The present study aims to investigate the function and the related mechanism by which HZ08 sensitizes radiotherapeutic efficiency to treat aggressive PCa cells. METHODS: PCa cells were pretreated with HZ08 (6,7-dimethoxy-1-(3,4-dimethoxy) benzyl-2-(N-n-octyl-N'-cyano) guanyl-1,2,3,4-tetrahydroisoquinoline) and followed by ionizing radiation (IR) treatment. Cytotoxicity in the treated cells was analyzed to assess the radiosensitization capacity of HZ08 by flow cytometry, MTT and colony survival assays. The cellular levels of reactive oxygen species (ROS) and oxygen consumption rates (OCR) were measured using specific ROS detection probes and a Seahorse XF96 Analyzer, respectively. RelB binding to the NF-κB intronic enhancer region of the human SOD2 gene was determined using a ChIP assay. The levels of phosphorylation of PI3K, Akt and IKKα were quantified and further confirmed using a PI3K inhibitor. Finally, the synergistic effect of HZ08 on radiosensitization of PCa cells was validated using a mouse xenograft tumor model. RESULTS: HZ08 enhanced radiosensitivity of PCa cells through increasing ROS and declining mitochondrial respiration due to suppression of mitochondrial antioxidant enzyme MnSOD. Mechanistically, HZ08 appeared to inhibit PI3K/Akt/IKKα signaling axis, resulting in transcriptional repression of MnSOD expression by preventing RelB nuclear translocation. CONCLUSIONS: HZ08 can serve as a useful radiosensitizing agent to improve radiotherapy for treating aggressive PCa cells with high level of constitutive RelB. The present study suggests a promising approach for enhancing radiotherapeutic efficiency to treat advanced PCa by inhibiting antioxidant defense function.


Subject(s)
Isoquinolines/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Superoxide Dismutase/biosynthesis , Transcription Factor RelB/metabolism , Animals , Cell Line, Tumor , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Oncogene Protein v-akt/antagonists & inhibitors , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Radiation Tolerance , Radiation-Sensitizing Agents/pharmacology , Random Allocation , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/metabolism , Transcription Factor RelB/antagonists & inhibitors , Xenograft Model Antitumor Assays
6.
Exp Cell Res ; 369(1): 129-138, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29778754

ABSTRACT

The subtypes of distant-organ metastasis led to treatment failure and poor prognosis are major obstacles in the management of patients with advanced breast cancer (BCa). Emerging evidences demonstrated that exosomes act as mediators for intercellular communication between various types of cells in the local tumor microenvironment. The present study aims to investigate whether BCa-derived exosomes are capable of cell-cell transferring miR-222 for BCa metastatic progression. Results showed that exosomal miR-222 is highly expressed in BCa patients with lymphatic metastasis. Consistently, the elevated levels of exosomal miR-222 are closely correlated with the high aggressivity of BCa cell lines. miR-222 promoting the aggressivity of BCa cells was confirmed in vitro and in vivo. Mechanistically, miR-222 directly targets PDLIM2, a tumor suppressor gene, leading to activation of NF-κB signal pathway. In conclusion, the levels of exosomal miR-222 are correlated with BCa metastatic progression. Exosome-transferred miR-222 promotes migration and invasion of BCa cells. miR-222 contributes to tumorigenicity of BCa cells through down-regulation of PDLIM2 and consequently activating NF-κB.


Subject(s)
Breast Neoplasms/pathology , Exosomes/metabolism , MicroRNAs/metabolism , RNA Transport/physiology , Transcription Factor RelA/physiology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Case-Control Studies , Cell Movement/genetics , Cells, Cultured , Exosomes/physiology , Female , Gene Expression Regulation, Neoplastic , Humans , LIM Domain Proteins/genetics , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Microfilament Proteins/genetics , Neoplasm Metastasis , Signal Transduction/genetics , Transcription Factor RelA/genetics , Tumor Microenvironment/physiology
7.
Cytokine ; 108: 151-159, 2018 08.
Article in English | MEDLINE | ID: mdl-29609137

ABSTRACT

Therapeutic resistance leading to tumor relapse is a major challenge in breast cancer (BCa) treatment. Numerous factors involved in multiple mechanisms promote the development of tumor chemo/radio-resistance. Cytokines/chemokines are important inflammatory factors and highly related to tumorigenesis, metastasis and tumors responses to treatment. A large number of studies have demonstrated that the network of cytokines activates multiple cell signaling pathways to promote tumor cell survival, proliferation, invasion, and migration. Particularly in BCa, cytokines-enhanced the epithelial-mesenchymal transition (EMT) process plays a pivotal role in the progression of metastatic phenotypes and resistance to the traditional chemo/radio-therapy. Virtually, therapeutic resistance is not entirely determined by tumor cell intrinsic characteristics but also dependent upon synchronized effects by numerous of local microenvironmental factors. Emerging evidence highlighted that exosomes secreted from various types of cells promote intercellular communication by transferring bioactive molecules including miRNAs and cytokines, suggesting that exosomes are essential for sustentation of tumor progression and therapeutic resistance within the tumor microenvironment. In this review, we discuss the mechanisms by which cytokines promote therapeutic resistance of BCa and suggest a potential approach for improving BCa therapeutics by inhibition of exosome function.


Subject(s)
Breast Neoplasms/drug therapy , Cytokines/immunology , Drug Resistance, Neoplasm , Exosomes/immunology , Tumor Microenvironment/immunology , Animals , Breast Neoplasms/immunology , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , Humans , Mice , MicroRNAs , Neoplasm Recurrence, Local/immunology , Signal Transduction
8.
Onco Targets Ther ; 11: 1529-1541, 2018.
Article in English | MEDLINE | ID: mdl-29593419

ABSTRACT

Owing to improved early surveillance and advanced therapy strategies, the current death rate due to breast cancer has decreased; nevertheless, drug resistance and relapse remain obstacles on the path to successful systematic treatment. Multiple mechanisms responsible for drug resistance have been elucidated, and miRNAs seem to play a major part in almost every aspect of cancer progression, including tumorigenesis, metastasis, and drug resistance. In recent years, exosomes have emerged as novel modes of intercellular signaling vehicles, initiating cell-cell communication through their fusion with target cell membranes, delivering functional molecules including miRNAs and proteins. This review particularly focuses on enumerating functional miRNAs involved in breast cancer drug resistance as well as their targets and related mechanisms. Subsequently, we discuss the prospects and challenges of miRNA function in drug resistance and highlight valuable approaches for the investigation of the role of exosomal miRNAs in breast cancer progression and drug resistance.

SELECTION OF CITATIONS
SEARCH DETAIL