Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Sci Rep ; 7(1): 4471, 2017 06 30.
Article in English | MEDLINE | ID: mdl-28667313

ABSTRACT

Surface enhanced Raman scattering (SERS) nanoparticles are an attractive alternative to fluorescent probes for biological labeling because of their photostability and multiplexing capabilities. However, nanoparticle size, shape, and surface properties are known to affect nanoparticle-cell interactions. Other issues such as the formation of a protein corona and antibody multivalency interfere with the labeling properties of nanoparticle-antibody conjugates. Hence, it is important to consider these aspects in order to validate such conjugates for live cell imaging applications. Using SERS nanoparticles that target HER2 and CD44 in breast cancer cells, we demonstrate labeling of fixed cells with high specificity that correlates well with fluorescent labels. However, when labeling live cells to monitor surface biomarker expression and dynamics, the nanoparticles are rapidly uptaken by the cells and become compartmentalized into different cellular regions. This behavior is in stark contrast to that of fluorescent antibody conjugates. This study highlights the impact of nanoparticle internalization and trafficking on the ability to use SERS nanoparticle-antibody conjugates to monitor cell dynamics.


Subject(s)
Microscopy , Nanoparticles , Spectrum Analysis, Raman , Biomarkers , Cell Line, Tumor , Flow Cytometry , Fluorescent Antibody Technique , Fluorescent Dyes , Humans , Hyaluronan Receptors/metabolism , Microscopy/methods , Molecular Imaging/methods , Receptor, ErbB-2/metabolism , Spectrum Analysis, Raman/methods
2.
J Vis Exp ; (108): 53417, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26967001

ABSTRACT

Although mitochondria possess their own transcriptional machinery, merely 1% of mitochondrial proteins are synthesized inside the organelle. The nuclear-encoded proteins are transported into mitochondria guided by their mitochondria targeting sequences (MTS); however, a majority of mitochondrial localized proteins lack an identifiable MTS. Nevertheless, the fact that MTS can instruct proteins to go into the mitochondria provides a valuable tool for studying mitochondrial functions of normally nuclear and/or cytoplasmic proteins. We have recently identified the cell cycle kinase CyclinB1/Cdk1 complex in the mitochondria. To specifically study the mitochondrial functions of this complex, mitochondrial overexpression and knock-down of this complex without interfering with its nuclear or cytoplasmic functions were essential. By tagging CyclinB1/Cdk1 with MTS, we were able to achieve mitochondrial overexpression of this complex to study its mitochondrial targets as well as functions. Via tagging dominant-negative Cdk1 with MTS, inhibition of Cdk1 activity was accomplished particularly in the mitochondria. Potential mitochondrial targets of CyclinB1/Cdk1 complex were identified using a gel-based proteomics approach. Unlike traditional 2D gel analysis, we employed 2-dimensional difference gel electrophoresis (2D-DIGE) technology followed by phosphoprotein staining to fluorescently label differentially phosphorylated proteins in mitochondrial Cdk1 expressing cells. Identification of phosphoprotein spots that were altered in wild type versus dominant negative Cdk1 bearing mitochondria revealed the identity of mitochondrial targets of Cdk1. Finally, to determine the effect of CyclinB1/Cdk1 mitochondrial localization in cell cycle progression, a cell proliferation assay using a synthetic thymidine analogue EdU (5-ethynyl-2'-deoxyuridine) was used to monitor the cells as they go through the cell cycle and replicate their DNA. Altogether, we demonstrated a variety of approaches available to study mitochondrial localization and activity of a cell cycle kinase. These are advanced, yet easy to follow methods that will be beneficial to many cell biology researchers.


Subject(s)
Mitochondria/enzymology , Mitochondrial Proteins/metabolism , CDC2 Protein Kinase/metabolism , CDC2 Protein Kinase/physiology , Cell Cycle/genetics , Cell Cycle/physiology , Cells, Cultured , Cyclin B1/metabolism , Electrophoresis, Gel, Two-Dimensional/methods , Humans , Mitochondrial Proteins/genetics , Phosphorylation
3.
Cell Rep ; 13(10): 2056-63, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26670043

ABSTRACT

Nuclear DNA repair capacity is a critical determinant of cell fate under genotoxic stress conditions. DNA repair is a well-defined energy-consuming process. However, it is unclear how DNA repair is fueled and whether mitochondrial energy production contributes to nuclear DNA repair. Here, we report a dynamic enhancement of oxygen consumption and mitochondrial ATP generation in irradiated normal cells, paralleled with increased mitochondrial relocation of the cell-cycle kinase CDK1 and nuclear DNA repair. The basal and radiation-induced mitochondrial ATP generation is reduced significantly in cells harboring CDK1 phosphorylation-deficient mutant complex I subunits. Similarly, mitochondrial ATP generation and nuclear DNA repair are also compromised severely in cells harboring mitochondrially targeted, kinase-deficient CDK1. These results demonstrate a mechanism governing the communication between mitochondria and the nucleus by which CDK1 boosts mitochondrial bioenergetics to meet the increased cellular fuel demand for DNA repair and cell survival under genotoxic stress conditions.


Subject(s)
Cyclin-Dependent Kinases/metabolism , DNA Damage/physiology , DNA Repair/physiology , Mitochondria/metabolism , Adenosine Triphosphate , Blotting, Western , CDC2 Protein Kinase , Cell Line , Comet Assay , DNA Damage/radiation effects , Energy Metabolism/physiology , Gene Knockdown Techniques , Humans , Protein Transport/radiation effects , RNA, Small Interfering , Radiation Effects , Transfection
4.
Mol Cancer Ther ; 14(9): 2090-102, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26141949

ABSTRACT

Tumor adaptive resistance to therapeutic radiation remains a barrier for further improvement of local cancer control. SIRT3, a member of the sirtuin family of NAD(+)-dependent protein deacetylases in mitochondria, promotes metabolic homeostasis through regulation of mitochondrial protein deacetylation and plays a key role in prevention of cell aging. Here, we demonstrate that SIRT3 expression is induced in an array of radiation-treated human tumor cells and their corresponding xenograft tumors, including colon cancer HCT-116, glioblastoma U87, and breast cancer MDA-MB231 cells. SIRT3 transcriptional activation is due to SIRT3 promoter activation controlled by the stress transcription factor NF-κB. Posttranscriptionally, SIRT3 enzymatic activity is further enhanced via Thr150/Ser159 phosphorylation by cyclin B1-CDK1, which is also induced by radiation and relocated to mitochondria together with SIRT3. Cells expressing Thr150Ala/Ser159Ala-mutant SIRT3 show a reduction in mitochondrial protein lysine deacetylation, Δψm, MnSOD activity, and mitochondrial ATP generation. The clonogenicity of Thr150Ala/Ser159Ala-mutant transfectants is lower and significantly decreased under radiation. Tumors harboring Thr150Ala/Ser159Ala-mutant SIRT3 show inhibited growth and increased sensitivity to in vivo local irradiation. These results demonstrate that enhanced SIRT3 transcription and posttranslational modifications in mitochondria contribute to adaptive radioresistance in tumor cells. CDK1-mediated SIRT3 phosphorylation is a potential effective target to sensitize tumor cells to radiotherapy.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Mitochondria/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Radiation Tolerance/genetics , Sirtuin 3/genetics , Transcriptional Activation , Acetylation , Animals , CDC2 Protein Kinase , Cell Line, Tumor , Disease Models, Animal , Enzyme Activation , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Mitochondria/radiation effects , Mitochondrial Proteins/metabolism , Mutation , NF-kappa B/metabolism , Neoplasms/pathology , Neoplasms/radiotherapy , Phosphorylation , Sirtuin 3/metabolism , Transcription, Genetic
5.
PLoS One ; 10(3): e0121046, 2015.
Article in English | MEDLINE | ID: mdl-25807077

ABSTRACT

A unique feature of cancer cells is to convert glucose into lactate to produce cellular energy, even under the presence of oxygen. Called aerobic glycolysis [The Warburg Effect] it has been extensively studied and the concept of aerobic glycolysis in tumor cells is generally accepted. However, it is not clear if aerobic glycolysis in tumor cells is fixed, or can be reversed, especially under therapeutic stress conditions. Here, we report that mTOR, a critical regulator in cell proliferation, can be relocated to mitochondria, and as a result, enhances oxidative phosphorylation and reduces glycolysis. Three tumor cell lines (breast cancer MCF-7, colon cancer HCT116 and glioblastoma U87) showed a quick relocation of mTOR to mitochondria after irradiation with a single dose 5 Gy, which was companied with decreased lactate production, increased mitochondrial ATP generation and oxygen consumption. Inhibition of mTOR by rapamycin blocked radiation-induced mTOR mitochondrial relocation and the shift of glycolysis to mitochondrial respiration, and reduced the clonogenic survival. In irradiated cells, mTOR formed a complex with Hexokinase II [HK II], a key mitochondrial protein in regulation of glycolysis, causing reduced HK II enzymatic activity. These results support a novel mechanism by which tumor cells can quickly adapt to genotoxic conditions via mTOR-mediated reprogramming of bioenergetics from predominantly aerobic glycolysis to mitochondrial oxidative phosphorylation. Such a "waking-up" pathway for mitochondrial bioenergetics demonstrates a flexible feature in the energy metabolism of cancer cells, and may be required for additional cellular energy consumption for damage repair and survival. Thus, the reversible cellular energy metabolisms should be considered in blocking tumor metabolism and may be targeted to sensitize them in anti-cancer therapy.


Subject(s)
Hexokinase/antagonists & inhibitors , Hexokinase/metabolism , Mitochondria/metabolism , Oxidative Phosphorylation/radiation effects , TOR Serine-Threonine Kinases/metabolism , Adenosine Triphosphate/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/radiotherapy , Cell Line, Tumor , Cell Proliferation/radiation effects , Energy Metabolism/radiation effects , Female , Glucose/metabolism , Glycolysis/radiation effects , HCT116 Cells , Humans , MCF-7 Cells , Mitochondria/radiation effects , Oxygen Consumption/radiation effects , Radiation
6.
Free Radic Biol Med ; 81: 77-87, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25578653

ABSTRACT

Mammalian cells are able to sense environmental oxidative and genotoxic conditions such as the environmental low-dose ionizing radiation (LDIR) present naturally on the earth's surface. The stressed cells then can induce a so-called radioadaptive response with an enhanced cellular homeostasis and repair capacity against subsequent similar genotoxic conditions such as a high dose radiation. Manganese superoxide dismutase (MnSOD), a primary mitochondrial antioxidant in mammals, has long been known to play a crucial role in radioadaptive protection by detoxifying O2(•-) generated by mitochondrial oxidative phosphorylation. In contrast to the well-studied mechanisms of SOD2 gene regulation, the mechanisms underlying posttranslational regulation of MnSOD for radioprotection remain to be defined. Herein, we demonstrate that cyclin D1/cyclin-dependent kinase 4 (CDK4) serves as the messenger to deliver the stress signal to mitochondria to boost mitochondrial homeostasis in human skin keratinocytes under LDIR-adaptive radioprotection. Cyclin D1/CDK4 relocates to mitochondria at the same time as MnSOD enzymatic activation peaks without significant changes in total MnSOD protein level. The mitochondrial-localized CDK4 directly phosphorylates MnSOD at serine-106 (S106), causing enhanced MnSOD enzymatic activity and mitochondrial respiration. Expression of mitochondria-targeted dominant negative CDK4 or the MnSOD-S106 mutant reverses LDIR-induced mitochondrial enhancement and adaptive protection. The CDK4-mediated MnSOD activation and mitochondrial metabolism boost are also detected in skin tissues of mice receiving in vivo whole-body LDIR. These results demonstrate a unique CDK4-mediated mitochondrial communication that allows cells to sense environmental genotoxic stress and boost mitochondrial homeostasis by enhancing phosphorylation and activation of MnSOD.


Subject(s)
Cyclin-Dependent Kinase 4/genetics , Keratinocytes/radiation effects , Mitochondria/radiation effects , Superoxide Dismutase/genetics , Adaptation, Physiological , Animals , Cell Line , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , Dose-Response Relationship, Radiation , Gene Expression Regulation , Humans , Keratinocytes/cytology , Keratinocytes/enzymology , Membrane Potential, Mitochondrial/radiation effects , Mice , Mice, Inbred BALB C , Mitochondria/enzymology , Oxidative Phosphorylation , Phosphorylation/drug effects , Radiation Tolerance , Radiation, Ionizing , Signal Transduction , Superoxide Dismutase/metabolism , Whole-Body Irradiation
7.
Mol Cell Oncol ; 2(4): e997518, 2015.
Article in English | MEDLINE | ID: mdl-27308517

ABSTRACT

Mitogen-activated protein kinase phosphatase 1 (MKP1 or DUSP1) is an antiapoptotic phosphatase that is overexpressed in many cancers, including breast cancer. MKP1 expression is inducible in radiation-treated breast cancer cells, and correlates with human epidermal growth factor receptor 2 (ERBB2, HER2) expression. The role of MKP1 in therapy resistance suggests that targeting MKP1 in HER2-positive breast tumors may significantly enhance the efficacy of anti-HER2 and other anticancer therapies.

8.
Cancer Res ; 74(24): 7498-509, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25377473

ABSTRACT

The MAPK phosphatase MKP1 (DUSP1) is overexpressed in many human cancers, including chemoresistant and radioresistant breast cancer cells, but its functional contributions in these settings are unclear. Here, we report that after cell irradiation, MKP1 translocates into mitochondria, where it prevents apoptotic induction by limiting accumulation of phosphorylated active forms of the stress kinase JNK. Increased levels of mitochondrial MKP1 after irradiation occurred in the mitochondrial inner membrane space. Notably, cell survival regulated by mitochondrial MKP1 was responsible for conferring radioresistance in HER2-overexpressing breast cancer cells, due to the fact that MKP1 serves as a major downstream effector in the HER2-activated RAF-MEK-ERK pathway. Clinically, we documented MKP1 expression exclusively in HER2-positive breast tumors, relative to normal adjacent tissue from the same patients. MKP1 overexpression was also detected in irradiated HER2-positive breast cancer stem-like cells (HER2(+)/CD44(+)/CD24(-/low)) isolated from a radioresistant breast cancer cell population after long-term radiation treatment. MKP1 silencing reduced clonogenic survival and enhanced radiosensitivity in these stem-like cells. Combined inhibition of MKP1 and HER2 enhanced cell killing in breast cancer. Together, our findings identify a new mechanism of resistance in breast tumors and reveal MKP1 as a novel therapeutic target for radiosensitization.


Subject(s)
Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Dual Specificity Phosphatase 1/biosynthesis , Radiation Tolerance/genetics , Receptor, ErbB-2/genetics , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Dual Specificity Phosphatase 1/genetics , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , MAP Kinase Kinase 4/metabolism , MCF-7 Cells , Membrane Potential, Mitochondrial/radiation effects , Mitochondria/genetics , Mitochondria/radiation effects , Protein Transport/radiation effects
9.
Dev Cell ; 29(2): 217-32, 2014 Apr 28.
Article in English | MEDLINE | ID: mdl-24746669

ABSTRACT

A substantial amount of mitochondrial energy is required for cell-cycle progression. The mechanisms underlying the coordination of the mitochondrial respiration with cell-cycle progression, especially the G2/M transition, remain to be elucidated. Here, we show that a fraction of cyclin B1/Cdk1 proteins localizes to the matrix of mitochondria and phosphorylates a cluster of mitochondrial proteins, including the complex I (CI) subunits in the respiratory chain. Cyclin B1/Cdk1-mediated CI phosphorylation enhances CI activity, whereas deficiency of such phosphorylation in each of the relevant CI subunits results in impairment of CI function. Mitochondria-targeted cyclin B1/Cdk1 increases mitochondrial respiration with enhanced oxygen consumption and ATP generation, which provides cells with efficient bioenergy for G2/M transition and shortens overall cell-cycle time. Thus, cyclin B1/Cdk1-mediated phosphorylation of mitochondrial substrates allows cells to sense and respond to increased energy demand for G2/M transition and, subsequently, to upregulate mitochondrial respiration for successful cell-cycle progression.


Subject(s)
Cell Division/physiology , Cyclin B1/metabolism , Cyclin-Dependent Kinases/metabolism , G2 Phase/physiology , Mitochondria/metabolism , Animals , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cyclin B1/genetics , Cyclin-Dependent Kinases/genetics , Electron Transport/physiology , Epithelial Cells/cytology , Humans , Keratinocytes/cytology , Liver/cytology , MCF-7 Cells , Mice , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitosis/physiology , Phosphorylation/physiology , Substrate Specificity/physiology , Cyclin-Dependent Kinase-Activating Kinase
10.
J Cancer Res Clin Oncol ; 140(1): 1-14, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23990015

ABSTRACT

PURPOSE: The lethal effects of cancer are associated with the enhanced tumor aggressiveness in recurrent and metastatic lesions that show resistant phenotype to anti-cancer therapy, a major barrier to improving overall survival of cancer patients. The presence of heterogeneous populations of cancer cells within a specific tumor including the tumor-initiating cells or so-called cancer stem cells (CSCs) has linked the acquired resistance (AR, or adaptive resistance). Herein, we discuss the CSC-mediated tumor repopulation in AR of breast cancer in this review. METHODS: We emphasize a dynamic feature of gene induction in tumor cells that undergo long-term treatment, and describe a specific HER2-NF-κB-HER2 pro-survival pathway that can be initiated in breast CSCs upon radiation therapy. RESULTS: Elucidation of HER2-induced pro-survival networks, specifically the force driving tumor repopulation due to radioresistant CSCs during anticancer therapies, will have a significant impact on the generation of new diagnostic and therapeutic targets to control of recurrent and metastatic breast tumors.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Neoplastic Stem Cells/pathology , Receptor, ErbB-2/metabolism , Adaptation, Biological , Animals , Female , Humans , NF-kappa B/metabolism , Receptor, ErbB-2/genetics
11.
Antioxid Redox Signal ; 20(10): 1599-617, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-23581847

ABSTRACT

SIGNIFICANCE: The mitochondrial antioxidant manganese superoxide dismutase (MnSOD) is encoded by genomic DNA and its dismutase function is fully activated in the mitochondria to detoxify free radical O2(•-) generated by mitochondrial respiration. Accumulating evidence shows an extensive communication between the mitochondria and cytoplasm under oxidative stress. Not only is the MnSOD gene upregulated by oxidative stress, but MnSOD activity can be enhanced via the mitochondrial protein influx (MPI). RECENT ADVANCES: A cluster of MPI containing cytoplasmic/nuclear proteins, such as cyclins, cyclin-dependent kinases, and p53 interact with and alter MnSOD activity. These proteins modulate MnSOD superoxide scavenging activity via post-translational modifications in the mitochondria. In addition to well-established pathways in gene expression, recent findings suggest that MnSOD enzymatic activity can also be enhanced by phosphorylation of specific motifs in mitochondria. This review attempts to discuss the pre- and post-translational regulation of MnSOD, and how these modifications alter MnSOD activity, which induces a cell adaptive response to oxidative stress. CRITICAL ISSUES: MnSOD is biologically significant to aerobic cells. Its role in protecting the cells against the deleterious effects of reactive oxygen species is evident. However, the exact network of MnSOD-associated cellular adaptive reaction to oxidative stress and its post-translational modifications, especially its enzymatic enhancement via phosphorylation, is not yet fully understood. FUTURE DIRECTIONS: The broad discussion of the multiple aspects of MnSOD regulation, including gene expression, protein modifications, and enzymatic activity, will shed light onto the unknown mechanisms that govern the prosurvival networks involved in cellular and mitochondrial adaptive response to genotoxic environment.


Subject(s)
Mitochondrial Proteins/metabolism , Oxidative Stress , Superoxide Dismutase/physiology , Animals , Gene Expression Regulation , Humans , Protein Processing, Post-Translational , Protein Transport , Tumor Suppressor Protein p53/metabolism
12.
J Mol Cell Biol ; 5(3): 166-75, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23243068

ABSTRACT

Manganese superoxide dismutase (MnSOD), a major antioxidant enzyme within the mitochondria, is responsible for the detoxification of free radicals generated by cellular metabolism and environmental/therapeutic irradiation. Cell cycle-dependent kinase Cdk1, along with its regulatory partner CyclinB1, plays important roles in the regulation of cell cycle progression as well as in genotoxic stress response. Herein, we identified the presence of the minimal Cdk1 phosphorylation consensus sequence ([S/T]-P; Ser106) in human MnSOD, suggesting Cdk1 as a potential upstream kinase of MnSOD. A substantial amount of CyclinB1/Cdk1 was found to localize in the mitochondrion upon irradiation. The enhanced Cdk1/MnSOD interaction and MnSOD phosphorylation were detected in both the irradiated human cells and mouse tissues. We report that CyclinB1/Cdk1 can regulate MnSOD through reversible Ser106 phosphorylation, both in vivo and in vitro. The CyclinB1/Cdk1-mediated MnSOD Ser106 resulted in increased MnSOD activity and stability, along with improved mitochondrial function and cellular resistance to radiation-induced apoptosis. These results demonstrate a unique pro-survival mechanism by which cells enhance the survival via CyclinB1/Cdk1-mediated MnSOD activation under genotoxic stress conditions.


Subject(s)
CDC2 Protein Kinase/metabolism , Cyclin B1/metabolism , Mitochondria/radiation effects , Superoxide Dismutase/metabolism , Animals , Antioxidants/metabolism , Apoptosis/genetics , Apoptosis/radiation effects , Cell Cycle/genetics , Cell Cycle/radiation effects , Cell Line, Tumor , DNA Damage , Female , Humans , Mice , Mice, Inbred BALB C , Mitochondria/metabolism , Phosphorylation , Protein Transport
13.
Clin Cancer Res ; 18(24): 6634-47, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23091114

ABSTRACT

PURPOSE: To understand the role of HER2-associated signaling network in breast cancer stem cells (BCSC) using radioresistant breast cancer cells and clinical recurrent breast cancers to evaluate HER2-targeted therapy as a tumor eliminating strategy for recurrent HER2(-/low) breast cancers. EXPERIMENTAL DESIGN: HER2-expressing BCSCs (HER2(+)/CD44(+)/CD24(-/low)) were isolated from radiation-treated breast cancer MCF7 cells and in vivo irradiated MCF7 xenograft tumors. Tumor aggressiveness and radioresistance were analyzed by gap filling, Matrigel invasion, tumor-sphere formation, and clonogenic survival assays. The HER2/CD44 feature was analyzed in 40 primary and recurrent breast cancer specimens. Protein expression profiling in HER2(+)/CD44(+)/CD24(-/low) versus HER2(-)/CD44(+)/CD24(-/low) BCSCs was conducted with two-dimensional difference gel electrophoresis (2-D DIGE) and high-performance liquid chromatography tandem mass spectrometry (HPLC/MS-MS) analysis and HER2-mediated signaling network was generated by MetaCore program. RESULTS: Compared with HER2-negative BCSCs, HER2(+)/CD44(+)/CD24(-/low) cells showed elevated aldehyde dehydrogenase (ALDH) activity and aggressiveness tested by Matrigel invasion, tumor sphere formation, and in vivo tumorigenesis. The enhanced aggressive phenotype and radioresistance of the HER2(+)/CD44(+)/CD24(-/low) cells were markedly reduced by inhibition of HER2 via siRNA or Herceptin treatments. Clinical breast cancer specimens revealed that cells coexpressing HER2 and CD44 were more frequently detected in recurrent (84.6%) than primary tumors (57.1%). In addition, 2-D DIGE and HPLC/MS-MS of HER2(+)/CD44(+)/CD24(-/low) versus HER2(-)/CD44(+)/CD24(-/low) BCSCs reported a unique HER2-associated protein profile including effectors involved in tumor metastasis, apoptosis, mitochondrial function, and DNA repair. A specific feature of HER2-STAT3 network was identified. CONCLUSION: This study provides the evidence that HER2-mediated prosurvival signaling network is responsible for the aggressive phenotype of BCSCs that could be targeted to control the therapy-resistant HER2(-/low) breast cancer.


Subject(s)
Breast Neoplasms/radiotherapy , Neoplastic Stem Cells/radiation effects , Receptor, ErbB-2/metabolism , Animals , Apoptosis/radiation effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CD24 Antigen/metabolism , Cell Movement , Cell Transformation, Neoplastic/radiation effects , Female , Gene Expression , Humans , Hyaluronan Receptors/metabolism , MCF-7 Cells , Membrane Potential, Mitochondrial/radiation effects , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasm Recurrence, Local/metabolism , Neoplastic Stem Cells/metabolism , Radiation Tolerance , Receptor, ErbB-2/genetics , STAT3 Transcription Factor/metabolism , Spheroids, Cellular/metabolism , Transcriptional Activation , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...