Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Oncotarget ; 7(40): 65837-65848, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27589564

ABSTRACT

Lymphocyte antigen 6 complex, locus E (LY6E) has been implicated in the malignant progression of various types of cancers; however, the underlying mechanism remains unclear. Here, we identified LY6E as an activator of HIF-1 and revealed their mechanistic and functional links in malignant tumor growth. The aberrant overexpression of LY6E increased HIF-1α gene expression principally at the transcription level. This, in turn, led to the expression of the pro-angiogenic factors, VEGFA and PDGFB, through decreases in the expression levels of PTEN mRNA and subsequent activation of the PI3K/Akt pathway. The LY6E-HIF-1 axis functioned to increase tumor blood vessel density and promoted tumor growth in immunodeficient mice. LY6E expression levels were significantly higher in human breast cancers than in normal breast tissues, and were strongly associated with the poor prognoses of various cancer patients. Our results characterized LY6E as a novel conductor of tumor growth through its modulation of the PTEN/PI3K/Akt/HIF-1 axis and demonstrated the validity of targeting this pathway for cancer therapy.


Subject(s)
Antigens, Surface/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Antigens, Surface/genetics , Apoptosis , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Proliferation , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Prognosis , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Nat Commun ; 6: 6153, 2015 Jan 23.
Article in English | MEDLINE | ID: mdl-25615526

ABSTRACT

Hypoxia-inducible factor 1 (HIF-1) plays a role in tumour metastases; however, the genes that activate HIF-1 and subsequently promote metastases have yet to be identified. Here we show that Ubiquitin C-terminal hydrolase-L1 (UCHL1) abrogates the von Hippel-Lindau-mediated ubiquitination of HIF-1α, the regulatory subunit of HIF-1, and consequently promotes metastasis. The aberrant overexpression of UCHL1 facilitates distant tumour metastases in a HIF-1-dependent manner in murine models of pulmonary metastasis. Meanwhile, blockade of the UCHL1-HIF-1 axis suppresses the formation of metastatic tumours. The expression levels of UCHL1 correlate with those of HIF-1α and are strongly associated with the poor prognosis of breast and lung cancer patients. These results indicate that UCHL1 promotes metastases as a deubiquitinating enzyme for HIF-1α, which justifies exploiting it as a prognostic marker and therapeutic target of cancers.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms/diagnosis , Neoplasms/pathology , Ubiquitin Thiolesterase/metabolism , Ubiquitination , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Targeted Therapy , NIH 3T3 Cells , Neoplasm Metastasis , Neoplasms/metabolism , Prognosis , Protein Stability , Up-Regulation
3.
Sci Rep ; 4: 3793, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24452734

ABSTRACT

Hypoxia-inducible factor 1 (HIF-1) has been associated with distant tumor metastasis; however, its function in multiple metastatic processes has not yet been fully elucidated. In the present study, we demonstrated that cancer cells transiently upregulated HIF-1 activity during their metastatic colonization after extravasation in the lungs in hypoxia-independent and reactive oxygen species (ROS)-dependent manners. Transient activation induced the expression of lactate dehydrogenase A and phosphorylation of the E1α subunit of pyruvate dehydrogenase, which indicated the reprogramming of glucose metabolic pathways from mitochondrial oxidative phosphorylation to anaerobic glycolysis and lactic acid fermentation. The administration of the HIF-1 inhibitor, YC-1, inhibited this reprogramming, increased intratumoral ROS levels, and eventually suppressed the formation of metastatic lung tumors. These results indicate that HIF-1-mediated metabolic reprogramming is responsible for the survival of metastatic cancers during their colonization in lungs by reducing cytotoxic ROS levels; therefore, its blockade by HIF-1-inhibitors is a rational strategy to prevent tumor metastasis.


Subject(s)
Glucose/metabolism , Hypoxia-Inducible Factor 1/metabolism , Lactic Acid/metabolism , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Mitochondria/pathology , Reactive Oxygen Species/metabolism , Anaerobiosis , Animals , Female , Glycolysis , Immunoenzyme Techniques , Lung Neoplasms/metabolism , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Nude , Mitochondria/metabolism , Oxidative Phosphorylation , Signal Transduction
4.
Nat Commun ; 3: 783, 2012 Apr 17.
Article in English | MEDLINE | ID: mdl-22510688

ABSTRACT

Tumour recurrence frequently occurs after radiotherapy, but the characteristics, intratumoural localization and post-irradiation behaviour of radioresistant cancer cells remain largely unknown. Here we develop a sophisticated strategy to track the post-irradiation fate of the cells, which exist in perinecrotic regions at the time of radiation. Although the perinecrotic tumour cells are originally hypoxia-inducible factor 1 (HIF-1)-negative, they acquire HIF-1 activity after surviving radiation, which triggers their translocation towards tumour blood vessels. HIF-1 inhibitors suppress the translocation and decrease the incidence of post-irradiation tumour recurrence. For the first time, our data unveil the HIF-1-dependent cellular dynamics during post-irradiation tumour recurrence and provide a rational basis for targeting HIF-1 after radiation therapy.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Neoplasms/physiopathology , Neoplasms/radiotherapy , Animals , Cell Hypoxia/radiation effects , Cell Line, Tumor , Cell Movement/radiation effects , Cell Survival/radiation effects , Humans , Male , Mice , Mice, Inbred BALB C , Neoplasms/blood supply , Neoplasms/metabolism , Radiation Tolerance , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/metabolism , Xenograft Model Antitumor Assays
5.
J Control Release ; 144(1): 109-14, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20096316

ABSTRACT

Matrix metalloproteinases (MMPs) are endopeptidases responsible for degrading the extracellular matrix (ECM) and remodeling tissue in both physiological and pathological processes. MMP2 and membrane-type 1 MMP (MT1-MMP) have been associated with tumor invasion, metastasis and angiogenesis; therefore, a molecular imaging strategy assessing their activity may help to predict the malignancy of tumors. Here, we established a novel method of specifically tagging the surface of MMP2- and MT1-MMP-positive cells, and applied it to the development of an optical imaging probe. We constructed a protein-based probe composed of a glutathione-S-transferase (GST)-tag (Inhibitory [I]-domain), a polypeptide as a specific substrate for both MMP2 and MT1-MMP (Cleaved [C]-domain), a transmembrane domain of the epidermal growth factor receptor (Transmembrane [TM]-domain), and DsRed2 (Fluorescent [F]-domain). In vitro experiments clearly demonstrated that, after the probe was cleaved at the C-domain by the MMPs, the resultant TM-F-domain was inserted into the cellular membrane. Optical imaging experiments in vivo demonstrated that the probe was cleaved and specifically remained in tumor xenografts in a MMP-dependent manner. These results indicate that the release of the I-C-domain through the proteolytic cleavage of the C-domain by MMP2 and MT1-MMP triggers the tagging of cellular membranes with the TM-F-domain. The present feasibility study opens the door to the development of a novel imaging probe for tumor malignancy using positron emission tomography as well as an optical imaging device.


Subject(s)
Matrix Metalloproteinases/metabolism , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Cells/metabolism , Cellular Structures/metabolism , Diagnostic Imaging , Endopeptidases/metabolism , ErbB Receptors/metabolism , Extracellular Matrix/metabolism , HeLa Cells , Humans , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/metabolism , Membranes/metabolism , Mice , Mice, Nude , Molecular Imaging , Peptides , Skin Neoplasms
6.
J Biol Chem ; 284(8): 5332-42, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19098000

ABSTRACT

Transcriptional activity of HIF-1 (hypoxia-inducible factor-1) has been reported to be up-regulated in solid tumors after ionizing radiation; however, the molecular mechanism underlying the response remains to be elucidated. In the present study, we performed a series of molecular imaging experiments using a HIF-1-dependent reporter gene, 5HREp-ODD-luc, and found an essential role of the Akt/mTOR pathway. Hypoxic tumor cells distant from blood vessels were dramatically reoxygenated at 24 h postirradiation, and HIF-1 activity increased as HIF-1alpha accumulated in the reoxygenated regions. The accumulation was inhibited with a nonmetabolizable glucose analog, 2-deoxy-d-glucose, through the suppression of radiation-induced phosphorylation of Akt in the reoxygenated regions. Akt knockdown and an mTOR inhibitor revealed the importance of the Akt/mTOR pathway in the postirradiation accumulation of HIF-1alpha. In vitro experiments confirmed that an increase in glucose availability induced Akt phosphorylation under reoxygenated conditions and consequently up-regulated HIF-1alpha translation. Moreover, both the accelerated translation and the previously reported reactive oxygen species-mediated stabilization of HIF-1alpha protein were essential to the activation of HIF-1. All of these results indicate that Akt/mTOR-dependent translation of HIF-1alpha plays a critical role in the postirradiation up-regulation of intratumoral HIF-1 activity in response to radiation-induced alterations of glucose and oxygen availability in a solid tumor.


Subject(s)
Gamma Rays , Glucose/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Hypoxia/radiation effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/genetics , Neoplasms/pathology , Oxygen/metabolism , Oxygen Consumption/radiation effects , Protein Biosynthesis/radiation effects , Protein Kinases/genetics , Protein Stability/radiation effects , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases
7.
Cancer Sci ; 99(11): 2327-35, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18823375

ABSTRACT

The therapeutic effect of concurrent chemoradiotherapy with TS-1 has been confirmed in various solid tumors; however, the detailed mechanism of action has not yet been fully elucidated. In the present study, we identified hypoxia-inducible factor-1 (HIF-1) as one of the targets of TS-1 in chemoradiotherapy. In growth delay assays using a tumor xenograft of non-small-cell lung carcinoma, H441, TS-1 treatment enhanced the therapeutic effect of single gamma-ray radiotherapy (14 Gy) and significantly delayed tumor growth by 1.58-fold compared to radiotherapy alone (P < 0.01). An optical in vivo imaging experiment using a HIF-1-dependent 5HRE-luc reporter gene revealed that TS-1 treatment suppressed radiation-induced activation of HIF-1 in the tumor xenografts. The suppression led to apoptosis of endothelial cells resulting in both a significant decrease in microvessel density (P < 0.05; vs radiation therapy alone) and a significant increase in apoptosis of tumor cells (P < 0.01; vs radiation therapy alone) in tumor xenografts. All of these results indicate that TS-1 enhances radiation-induced apoptosis of endothelial cells by suppressing HIF-1 activity, resulting in an increase in radiosensitivity of the tumor cells. Our findings strengthen the importance of both HIF-1 and its downstream gene, such as vascular endothelial cell growth factor, as therapeutic targets to enhance the effect of radiotherapy.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Apoptosis , Carcinoma, Non-Small-Cell Lung/therapy , Hypoxia-Inducible Factor 1/metabolism , Lung Neoplasms/therapy , Oxonic Acid/therapeutic use , Tegafur/therapeutic use , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Line, Tumor , Combined Modality Therapy , Drug Combinations , Endothelial Cells/metabolism , Endothelial Cells/radiation effects , Humans , Immunohistochemistry , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Mice , Mice, Inbred BALB C , Vascular Endothelial Growth Factor A/metabolism
8.
Biochem Biophys Res Commun ; 373(4): 533-8, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18586004

ABSTRACT

As the transcriptional activity of hypoxia-inducible factor 1 (HIF-1) is associated with resistance of tumor cells to current antitumor therapies, the spatiotemporal dynamics of HIF-1-active regions has been of great interest as a therapeutic target. In the present study, we established a unique cancer cell line, which changes color HIF-1-dependently, and monitored it during tumor progression. In imaging experiments, HIF-1-active cells appeared over the tumor xenograft, but dramatically decreased in number as blood vessels developed around the tumor. The remaining HIF-1-active cells at the center of the xenograft also disappeared after neovascularization. Thereafter, tumor growth was accelerated and HIF-1-active cells reappeared in different regions. The distance between HIF-1-active cells and the nearest vessels correlated to the diameter of the vessel (r=0.801). These results provide a basic knowledge of how to estimate the spatiotemporal dynamics of HIF-1-active cells using information about the image-guided architecture of tumor blood vessels.


Subject(s)
Cell Line, Tumor , Hypoxia-Inducible Factor 1/metabolism , Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Animals , HeLa Cells , Humans , Luminescent Proteins/analysis , Mice , Neoplasm Transplantation , Neoplasms/drug therapy , Xenograft Model Antitumor Assays
9.
Biochem Biophys Res Commun ; 360(4): 791-6, 2007 Sep 07.
Article in English | MEDLINE | ID: mdl-17624305

ABSTRACT

The transcriptional activity of hypoxia-inducible factor-1 (HIF-1) is associated with tumor malignancies; therefore, it is important to comprehend its dynamism in solid tumors. However, a molecular imaging strategy to accurately access it remains to be developed. We constructed here a novel HIF-1-dependent reporter gene, 5HREp-ODD-luc, in which 5 copies of the hypoxia-response element (5HRE) enhance expression of the oxygen-dependent degradation (ODD) domain and luciferase (luc) fusion under hypoxia. Because the ODD domain caused the oxygen-dependent degradation of the ODD-Luc protein, the novel reporter gene showed little leak of luminescence under normoxia. Such a property caused an increase of the hypoxia-responsiveness up to about 4.7 x 10(4) -fold. Moreover, the ODD domain caused rapid degradation of the ODD-Luc protein under normoxia, the luminescence reflected the dynamism of HIF-1 activity in real-time. The superiority of the novel reporter gene will surely accelerate analysis of the intratumoral HIF-1 activity during tumor progression and cancer treatments.


Subject(s)
Cell Hypoxia/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms/metabolism , Oxygen/metabolism , Base Sequence , DNA Primers , Genes, Reporter , HeLa Cells , Humans , Hydrolysis , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...