Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Sci Rep ; 8(1): 16932, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30446677

ABSTRACT

Ras GTPases are powerful drivers for tumorigenesis, but directly targeting Ras for treating cancer remains challenging. The growth and transforming activity of the aggressive basal-like breast cancer (BLBC) are driven by N-Ras. To target N-Ras in BLBC, this study screened existing pharmacologically active compounds for the new ability to induce N-Ras degradation, which led to the identification of flunarizine (FLN), previously approved for treating migraine and epilepsy. The FLN-induced N-Ras degradation was not affected by a 26S-proteasome inhibitor. Rather, it was blocked by autophagy inhibitors. Furthermore, N-Ras can be seen co-localized with active autophagosomes upon FLN treatment, suggesting that FLN alters the autophagy pathway to degrade N-Ras. Importantly, FLN treatment recapitulated the effect of N-RAS silencing in vitro by selectively inhibiting the growth of BLBC cells, but not that of breast cancer cells of other subtypes. In addition, in vivo FLN inhibited tumor growth of a BLBC xenograft model. In conclusion, this proof-of-principle study presents evidence that the autophagy pathway can be coerced by small molecule inhibitors, such as FLN, to degrade Ras as a strategy to treat cancer. FLN has low toxicity and should be further investigated to enrich the toolbox of cancer therapeutics.


Subject(s)
Autophagy/drug effects , Flunarizine/pharmacology , ras Proteins/metabolism , Animals , Autophagosomes , Autophagy/genetics , Cell Line, Tumor , Disease Models, Animal , Drug Screening Assays, Antitumor , Genes, Reporter , Humans , Mice , Proteolysis , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , ras Proteins/genetics
2.
Sci Transl Med ; 10(464)2018 10 24.
Article in English | MEDLINE | ID: mdl-30355798

ABSTRACT

Medulloblastoma (MB) is the most common malignant brain tumor of childhood. Although outcomes have improved in recent decades, new treatments are still needed to improve survival and reduce treatment-related complications. The MB subtypes groups 3 and 4 represent a particular challenge due to their intragroup heterogeneity, which limits the options for "rational" targeted therapies. Here, we report a systems biology approach to drug repositioning that integrates a nonparametric, bootstrapping-based simulated annealing algorithm and a 3D drug functional network to characterize dysregulated driver signaling networks, thereby identifying potential drug candidates. From more than 1300 drug candidates studied, we identified five members of the cardiac glycoside family as potentially inhibiting the growth of groups 3 and 4 MB and subsequently confirmed this in vitro. Systemic in vivo treatment of orthotopic patient-derived xenograft (PDX) models of groups 3 and 4 MB with digoxin, a member of the cardiac glycoside family approved for the treatment of heart failure, prolonged animal survival at plasma concentrations known to be tolerated in humans. These results demonstrate the power of a systematic drug repositioning method in identifying a potential treatment for MB. Our strategy could potentially be used to accelerate the repositioning of treatments for other human cancers that lack clearly defined rational targets.


Subject(s)
Brain Neoplasms/drug therapy , Digoxin/therapeutic use , Drug Repositioning , Medulloblastoma/drug therapy , Systems Biology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Brain Neoplasms/blood , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Digoxin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Medulloblastoma/blood , Medulloblastoma/genetics , Mice, Inbred NOD , Mice, SCID , Mitochondria/drug effects , Mitochondria/metabolism , Radiation, Ionizing , Signal Transduction/drug effects , Signal Transduction/radiation effects , Survival Analysis , Xenograft Model Antitumor Assays
3.
Cancer Res ; 78(8): 2052-2064, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29567857

ABSTRACT

Triple-negative breast cancer (TNBC) exhibits more traits possessed by cancer stem cells (CSC) than other breast cancer subtypes and is more likely to develop brain metastases. TNBC patients usually have shorter survival time after diagnosis of brain metastasis, suggesting an innate ability of TNBC tumor cells in adapting to the brain. In this study, we establish novel animal models to investigate early tumor adaptation in brain metastases by introducing both patient-derived and cell line-derived CSC-enriched brain metastasis tumorsphere cells into mice. We discovered astrocyte-involved tumor activation of protocadherin 7 (PCDH7)-PLCß-Ca2+-CaMKII/S100A4 signaling as a mediator of brain metastatic tumor outgrowth. We further identified and evaluated the efficacy of a known drug, the selective PLC inhibitor edelfosine, in suppressing the PCDH7 signaling pathway to prohibit brain metastases in the animal models. The results of this study reveal a novel signaling pathway for brain metastases in TNBC and indicate a promising strategy of metastatic breast cancer prevention and treatment by targeting organ-adaptive cancer stem cells.Significance: These findings identify a compound to block adaptive signaling between cancer stem cells and brain astrocytes. Cancer Res; 78(8); 2052-64. ©2018 AACR.


Subject(s)
Adaptation, Physiological , Brain Neoplasms/prevention & control , Brain Neoplasms/secondary , Neoplastic Stem Cells/pathology , Triple Negative Breast Neoplasms/pathology , Animals , Cadherins/genetics , Cadherins/metabolism , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Line, Tumor , Cell Proliferation , Enzyme Inhibitors/pharmacology , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Phospholipase C beta/antagonists & inhibitors , Phospholipase C beta/metabolism , Phospholipid Ethers/pharmacology , Protocadherins , RNA, Messenger/genetics , Retrospective Studies , S100 Calcium-Binding Protein A4/metabolism , Signal Transduction
4.
Cancer Res ; 76(8): 2094-2104, 2016 04 15.
Article in English | MEDLINE | ID: mdl-26893478

ABSTRACT

The activation of the epithelial-to-mesenchymal transition (EMT) program is a critical step in cancer progression and metastasis, but visualization of this process at the single-cell level, especially in vivo, remains challenging. We established an in vivo approach to track the fate of tumor cells based on a novel EMT-driven fluorescent color switching breast cancer mouse model and intravital two-photon laser scanning microscopy. Specifically, the MMTV-PyMT, Rosa26-RFP-GFP, and Fsp1-Cre triple transgenic mouse model was used to monitor the conversion of RFP-positive epithelial cells to GFP-positive mesenchymal cells in mammary tumors under the control of the Fsp1 (ATL1) promoter, a gate-keeper of EMT initiation. RFP-positive cells were isolated from the tumors, sorted, and transplanted into mammary fat pads of SCID mice to monitor EMT during breast tumor formation. We found that the conversion from RFP- to GFP-positive and spindle-shaped cells was a gradual process, and that GFP-positive cells preferentially localized close to blood vessels, independent of tumor size. Furthermore, cells undergoing EMT expressed high levels of the HGF receptor, c-Met, and treatment of RFP-positive cells with the c-Met inhibitor, cabozantinib, suppressed the RFP-to-GFP conversion in vitro Moreover, administration of cabozantinib to mice with palpable RFP-positive tumors resulted in a silent EMT phenotype whereby GFP-positive cells exhibited reduced motility, leading to suppressed tumor growth. In conclusion, our imaging technique provides a novel opportunity for visualizing tumor EMT at the single-cell level and may help to reveal the intricacies underlying tumor dynamics and treatment responses. Cancer Res; 76(8); 2094-104. ©2016 AACR.


Subject(s)
Epithelial-Mesenchymal Transition , Mammary Neoplasms, Experimental/pathology , Animals , Disease Models, Animal , Female , Mammary Neoplasms, Experimental/diagnostic imaging , Mice , Mice, SCID , Mice, Transgenic , Microscopy, Fluorescence/methods
5.
Lab Chip ; 15(20): 4032-43, 2015 Oct 21.
Article in English | MEDLINE | ID: mdl-26329655

ABSTRACT

A truly practical lab-on-a-chip (LOC) system for point-of-care testing (POCT) hepatotoxicity assessment necessitates the embodiment of full-automation, ease-of-use and "sample-in-answer-out" diagnostic capabilities. To date, the reported microfluidic devices for POCT hepatotoxicity assessment remain rudimentary as they largely embody only semi-quantitative or single sample/gene detection capabilities. In this paper, we describe, for the first time, an integrated LOC system that is somewhat close to a practical POCT hepatotoxicity assessment device - it embodies both tissue sample preparation and multiplex real-time RT-PCR. It features semi-automation, is relatively easy to use, and has "sample-in-answer-out" capabilities for multiplex gene expression analysis. Our tissue sample preparation module incorporating both a microhomogenizer and surface-treated paramagnetic microbeads yielded high purity mRNA extracts, considerably better than manual means of extraction. A primer preloading surface treatment procedure and the single-loading inlet on our multiplex real-time RT-PCR module simplify off-chip handling procedures for ease-of-use. To demonstrate the efficacy of our LOC system for POCT hepatotoxicity assessment, we perform a preclinical animal study with the administration of cyclophosphamide, followed by gene expression analysis of two critical protein biomarkers for liver function tests, aspartate transaminase (AST) and alanine transaminase (ALT). Our experimental results depict normalized fold changes of 1.62 and 1.31 for AST and ALT, respectively, illustrating up-regulations in their expression levels and hence validating their selection as critical genes of interest. In short, we illustrate the feasibility of multiplex gene expression analysis in an integrated LOC system as a viable POCT means for hepatotoxicity assessment.


Subject(s)
Analytic Sample Preparation Methods/instrumentation , Gene Expression Regulation , Lab-On-A-Chip Devices , Liver/drug effects , Point-of-Care Systems , Real-Time Polymerase Chain Reaction/instrumentation , Toxicity Tests/instrumentation , Alanine Transaminase/genetics , Animals , Aspartate Aminotransferases/genetics , DNA Primers/genetics , Liver/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , RNA, Messenger/metabolism , Systems Integration
6.
Oncotarget ; 6(28): 25320-38, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26318291

ABSTRACT

Interactions among tumor cells, stromal cells, and extracellular matrix compositions are mediated through cytokines during tumor progression. Our analysis of 132 known cytokines and growth factors in published clinical breast cohorts and our 84 patient-derived xenograft models revealed that the elevated connective tissue growth factor (CTGF) in tumor epithelial cells significantly correlated with poor clinical prognosis and outcomes. CTGF was able to induce tumor cell epithelial-mesenchymal transition (EMT), and promote stroma deposition of collagen I fibers to stimulate tumor growth and metastasis. This process was mediated through CTGF-tumor necrosis factor receptor I (TNFR1)-IκB autocrine signaling. Drug treatments targeting CTGF, TNFR1, and IκB signaling each prohibited the EMT and tumor progression.


Subject(s)
Autocrine Communication , Breast Neoplasms/metabolism , Collagen Type I/metabolism , Connective Tissue Growth Factor/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Stromal Cells/metabolism , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Line, Tumor , Cell Movement , Cell Proliferation , Connective Tissue Growth Factor/antagonists & inhibitors , Connective Tissue Growth Factor/genetics , Disease-Free Survival , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , I-kappa B Proteins/antagonists & inhibitors , I-kappa B Proteins/metabolism , Kaplan-Meier Estimate , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , RNA Interference , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Stromal Cells/drug effects , Stromal Cells/pathology , Time Factors , Transfection , Xenograft Model Antitumor Assays
7.
Life Sci ; 134: 68-72, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26037401

ABSTRACT

AIMS: Cerebral ischemia is one of the common diseases treated by electro-acupuncture (EA). Although the clinical efficacy has been widely affirmed, the mechanisms of action leading to the health benefits are not understood. In this study, the role of EA in modulating the lactate energy metabolism and lactate transportation was explored on the middle cerebral artery occlusion (MCAO) ischemic rat model. MAIN METHODS: Repeated EA treatments once daily for 7 days were applied to the MCAO rats and neurological function evaluation was performed. Brain tissues were harvested for lactate concentration examination, immunohistochemical staining, Western blot and qRT-PCR analyses for the expressions of lactate transporter (monocarboxylate transporter 1, MCT1) and glial fibrillary acidic protein (GFAP). KEY FINDINGS: The animal behavioral tests showed that the 7-day EA treatments significantly promoted the recovery of neurological deficits in the MCAO rats, which correlated with the enhanced lactate energy metabolism in the ischemic brain. In the cortical ischemic area of the MCAO rats, EA treatments led to the activation of astrocytes, and induced a further increase of lactate transporter (monocarboxylate transporter 1, MCT1) expression in astrocytes at both protein and mRNA levels. SIGNIFICANCE: Our results suggest that the EA treatments activated lactate metabolism in the resident astrocytes around the ischemic area and up-regulated the expression of MCT1 in these astrocytes which facilitated the transfer of intracellular lactate to extracellular domain to be utilized by injured neurons to improve the neurological deficit.


Subject(s)
Astrocytes/metabolism , Brain , Electroacupuncture , Monocarboxylic Acid Transporters/biosynthesis , Stroke , Symporters/biosynthesis , Up-Regulation , Animals , Astrocytes/pathology , Biological Transport, Active , Brain/metabolism , Brain/pathology , Glial Fibrillary Acidic Protein/biosynthesis , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/therapy , Lactic Acid/metabolism , Male , Rats , Rats, Wistar , Stroke/metabolism , Stroke/pathology , Stroke/therapy
8.
Sci Rep ; 3: 3467, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24322672

ABSTRACT

Convincing epidemiological data suggest an inverse association between cancer and neurodegeneration, including Alzheimer's disease (AD). Since both AD and cancer are characterized by abnormal, but opposing cellular behavior, i.e., increased cell death in AD while excessive cell growth occurs in cancer, this motivates us to initiate the study into unraveling the shared genes and cell signaling pathways linking AD and glioblastoma multiform (GBM). In this study, a comprehensive bioinformatics analysis on clinical microarray datasets of 1,091 GBM and 524 AD cohorts was performed. Significant genes and pathways were identified from the bioinformatics analyses - in particular ERK/MAPK signaling, up-regulated in GBM and Angiopoietin Signaling pathway, reciprocally up-regulated in AD - connecting GBM and AD (P < 0.001), were investigated in details for their roles in GBM growth in an AD environment. Our results showed that suppression of GBM growth in an AD background was mediated by the ERK-AKT-p21-cell cycle pathway and anti-angiogenesis pathway.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Signal Transduction , Transcription, Genetic , Amyloid beta-Peptides/metabolism , Animals , Astrocytes/metabolism , Case-Control Studies , Cell Line , Cell Movement/genetics , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation , Glioblastoma/pathology , Humans , MAP Kinase Signaling System , Mice , Models, Biological , Neovascularization, Pathologic/metabolism , Proto-Oncogene Proteins c-akt/metabolism
9.
Cancer Res ; 73(20): 6149-63, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24097821

ABSTRACT

A new type of signaling network element, called cancer signaling bridges (CSB), has been shown to have the potential for systematic and fast-tracked drug repositioning. On the basis of CSBs, we developed a computational model to derive specific downstream signaling pathways that reveal previously unknown target-disease connections and new mechanisms for specific cancer subtypes. The model enables us to reposition drugs based on available patient gene expression data. We applied this model to repurpose known or shelved drugs for brain, lung, and bone metastases of breast cancer with the hypothesis that cancer subtypes have their own specific signaling mechanisms. To test the hypothesis, we addressed specific CSBs for each metastasis that satisfy (i) CSB proteins are activated by the maximal number of enriched signaling pathways specific to a given metastasis, and (ii) CSB proteins are involved in the most differential expressed coding genes specific to each breast cancer metastasis. The identified signaling networks for the three types of breast cancer metastases contain 31, 15, and 18 proteins and are used to reposition 15, 9, and 2 drug candidates for the brain, lung, and bone metastases. We conducted both in vitro and in vivo preclinical experiments as well as analysis on patient tumor specimens to evaluate the targets and repositioned drugs. Of special note, we found that the Food and Drug Administration-approved drugs, sunitinib and dasatinib, prohibit brain metastases derived from breast cancer, addressing one particularly challenging aspect of this disease.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Drug Repositioning , Models, Biological , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Microarray Analysis , Neoplasm Metastasis , Signal Transduction
10.
J Neurosci Res ; 91(1): 128-37, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23115024

ABSTRACT

Aberrant expression of epidermal growth factor receptor (EGFR; ErbB1) and HER2 (ErbB2) tyrosine kinases frequently occurs in glioblastoma multiforme (GBM) patients and is considered to be associated with tumor malignancy and poor patient prognosis. In the present study, a dual EGFR and HER2 inhibitor (GW2974) was evaluated for its effects in GBM in vitro and in vivo. Results showed that low-concentration GW2974 inhibited GBM cell invasion, whereas a high concentration of the same compound counteracted this effect. Similar results were observed in an intracranial GBM xenograft model, in which, although both doses of GW2974 slowed tumor growth, no improvement in survival was observed in mice treated with high-dose GW2974, presumably because of the augmentation of tumor invasion. By protein microarray and Western blot analyses, the p38 mitogen-activated protein kinase (MAPK) pathway was found to be activated in GBM cells under high-concentration GW2974. Additionally, blockage of the p38 MAPK pathway reproduced the inhibitory effect of low-concentration GW2974 on cell invasion. These data suggest that the p38 MAPK pathway might contribute to the differential regulation of cell invasion by GW2974. Taken together, our results indicate that GW2974 exerts different effects in GBM depending on drug dosage. This offers a new perspective on the role of GW2974 in tumor progression, providing a potential strategy for GBM treatment based on precise prescription.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Quinazolines/administration & dosage , Animals , Blotting, Western , Brain Neoplasms/pathology , Cell Line, Tumor , Dose-Response Relationship, Drug , ErbB Receptors/antagonists & inhibitors , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Male , Mice , Mice, SCID , Neoplasm Invasiveness/pathology , Protein Array Analysis , Receptor, ErbB-2/antagonists & inhibitors , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL