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.
Pigment Cell Melanoma Res ; 32(6): 792-808, 2019 11.
Article in English | MEDLINE | ID: mdl-31207090

ABSTRACT

In response to the dynamic intra-tumor microenvironment, melanoma cells adopt distinct phenotypic states associated with differential expression of the microphthalmia-associated transcription factor (MITF). The response to hypoxia is driven by hypoxia-inducible transcription factors (HIFs) that reprogram metabolism and promote angiogenesis. HIF1α indirectly represses MITF that can activate HIF1α expression. Although HIF and MITF share a highly related DNA-binding specificity, it is unclear whether they co-regulate subset of target genes. Moreover, the genomewide impact of hypoxia on melanoma and whether melanoma cell lines representing different phenotypic states exhibit distinct hypoxic responses is unknown. Here we show that three different melanoma cell lines exhibit widely different hypoxia responses with only a core 23 genes regulated in common after 12 hr in hypoxia. Surprisingly, under hypoxia MITF is transiently up-regulated by HIF1α and co-regulates a subset of HIF targets including VEGFA. Significantly, we also show that MITF represses itself and also regulates SDHB to control the TCA cycle and suppress pseudo-hypoxia. Our results reveal a previously unsuspected role for MITF in metabolism and the network of factors underpinning the hypoxic response in melanoma.


Subject(s)
Citric Acid Cycle , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/metabolism , Tumor Hypoxia , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Melanoma/genetics , Neoplasm Invasiveness , Succinate Dehydrogenase/metabolism , Tumor Hypoxia/genetics , Up-Regulation/genetics
2.
Cancer Res ; 76(13): 3744-55, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27197160

ABSTRACT

Tumor hypoxia is associated clinically with therapeutic resistance and poor patient outcomes. One feature of tumor hypoxia is activated expression of carbonic anhydrase IX (CA9), a regulator of pH and tumor growth. In this study, we investigated the hypothesis that impeding the reuptake of bicarbonate produced extracellularly by CA9 could exacerbate the intracellular acidity produced by hypoxic conditions, perhaps compromising cell growth and viability as a result. In 8 of 10 cancer cell lines, we found that hypoxia induced the expression of at least one bicarbonate transporter. The most robust and frequent inductions were of the sodium-driven bicarbonate transporters SLC4A4 and SLC4A9, which rely upon both HIF1α and HIF2α activity for their expression. In cancer cell spheroids, SLC4A4 or SLC4A9 disruption by either genetic or pharmaceutical approaches acidified intracellular pH and reduced cell growth. Furthermore, treatment of spheroids with S0859, a small-molecule inhibitor of sodium-driven bicarbonate transporters, increased apoptosis in the cell lines tested. Finally, RNAi-mediated attenuation of SLC4A9 increased apoptosis in MDA-MB-231 breast cancer spheroids and dramatically reduced growth of MDA-MB-231 breast tumors or U87 gliomas in murine xenografts. Our findings suggest that disrupting pH homeostasis by blocking bicarbonate import might broadly relieve the common resistance of hypoxic tumors to anticancer therapy. Cancer Res; 76(13); 3744-55. ©2016 AACR.


Subject(s)
Bicarbonates/metabolism , Hypoxia/physiopathology , Neoplasms/metabolism , Neoplasms/prevention & control , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Apoptosis , Blotting, Western , Carbonic Anhydrase IX/genetics , Carbonic Anhydrase IX/metabolism , Cell Proliferation , Female , Humans , Hydrogen-Ion Concentration , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasms/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sodium-Bicarbonate Symporters/genetics , Sodium-Bicarbonate Symporters/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Oncotarget ; 6(23): 19413-27, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26305601

ABSTRACT

Carbonic anhydrase IX (CAIX) is strongly induced by hypoxia and its overexpression is associated with poor therapeutic outcome in cancer. Here, we report that hypoxia promotes tumour heterogeneity through the epigenetic regulation of CAIX. Based on hypoxic CAIX expression we identify and characterize two distinct populations of tumour cells, one that has inducible expression of CAIX and one that does not. The CAIX+ve population is enriched with cells expressing cancer stem cell markers and which have high self-renewal capacity. We show that differential CAIX expression is due to differences in chromatin structure. To further investigate the relationship between chromatin organization and hypoxic induction of CAIX expression we investigated the effect of JQ1 an inhibitor of BET bromodomain proteins and A366 a selective inhibitor of the H3K9 methyltransferase G9a/GLP. We identified that these drugs were able to modulate hypoxic CAIX expression induction. This further highlights the role of epigenetic modification in adaption to hypoxia and also in regulation of heterogeneity of cells within tumours. Interestingly, we identified that the two subpopulations show a differential sensitivity to HDAC inhibitors, NaBu or SAHA, with the CAIX positive showing greater sensitivity to treatment. We propose that drugs modulating chromatin regulation of expression may be used to reduce heterogeneity induced by hypoxia and could in combination have significant clinical consequences.


Subject(s)
Antigens, Neoplasm/biosynthesis , Carbonic Anhydrases/biosynthesis , Histone Deacetylase Inhibitors/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Animals , Antigens, Neoplasm/genetics , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Line, Tumor , Enzyme Induction , Female , HCT116 Cells , Heterografts , Humans , Isoenzymes/biosynthesis , MCF-7 Cells , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/pathology
4.
Clin Cancer Res ; 18(11): 3100-11, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22498007

ABSTRACT

PURPOSE: Bevacizumab, an anti-VEGFA antibody, inhibits the developing vasculature of tumors, but resistance is common. Antiangiogenic therapy induces hypoxia and we observed increased expression of hypoxia-regulated genes, including carbonic anhydrase IX (CAIX), in response to bevacizumab treatment in xenografts. CAIX expression correlates with poor prognosis in most tumor types and with worse outcome in bevacizumab-treated patients with metastatic colorectal cancer, malignant astrocytoma, and recurrent malignant glioma. EXPERIMENTAL DESIGN: We knocked down CAIX expression by short hairpin RNA in a colon cancer (HT29) and a glioblastoma (U87) cell line which have high hypoxic induction of CAIX and overexpressed CAIX in HCT116 cells which has low CAIX. We investigated the effect on growth rate in three-dimensional (3D) culture and in vivo, and examined the effect of CAIX knockdown in combination with bevacizumab. RESULTS: CAIX expression was associated with increased growth rate in spheroids and in vivo. Surprisingly, CAIX expression was associated with increased necrosis and apoptosis in vivo and in vitro. We found that acidity inhibits CAIX activity over the pH range found in tumors (pK = 6.84), and this may be the mechanism whereby excess acid self-limits the build-up of extracellular acid. Expression of another hypoxia inducible CA isoform, CAXII, was upregulated in 3D but not two-dimensional culture in response to CAIX knockdown. CAIX knockdown enhanced the effect of bevacizumab treatment, reducing tumor growth rate in vivo. CONCLUSION: This work provides evidence that inhibition of the hypoxic adaptation to antiangiogenic therapy enhances bevacizumab treatment and highlights the value of developing small molecules or antibodies which inhibit CAIX for combination therapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Antigens, Neoplasm/metabolism , Carbonic Anhydrases/metabolism , Cell Proliferation/drug effects , Animals , Antigens, Neoplasm/genetics , Bevacizumab , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Cell Line, Tumor , Female , Gene Knockdown Techniques , Glioblastoma/metabolism , HCT116 Cells , HT29 Cells , Humans , Hydrogen-Ion Concentration , Mice , Necrosis , Neoplasm Transplantation , Transfection , Transplantation, Heterologous
5.
Am J Pathol ; 176(5): 2477-89, 2010 May.
Article in English | MEDLINE | ID: mdl-20382705

ABSTRACT

Autophagy is a self-degradation mechanism by which cells recycle their own cytoplasmic constituents and dispose of excess or defective organelles after starvation and oxygen deprivation. An antibody to the microtubule-associated protein 1 light chain 3 (LC3A), recognizing both the soluble (LC3A-I) and the membrane-bound form (LC3A-II) of the protein, was used to detect autophagic activity in 102 breast carcinomas. Three distinct patterns were recognized: (1) diffuse cytoplasmic, (2) cytoplasmic/juxta-nuclear, and (3) "stone-like" pattern--dense, rounded, amorphous structures, 5 microm on average, typically enclosed within cytoplasmic vacuoles. The diffuse cytoplasmic pattern showed a direct association with estrogen and progesterone receptor expression. The juxta-nuclear pattern indicated a similar association with hormone receptors, an inverse association with tumor size, and a favorable prognosis. By contrast, an increased number of stone-like structures, probably representing an excessive autophagic response, was related to high-grade tumors and a less favorable outcome. Interestingly, 60 additional epithelial tumors of nonbreast origin disclosed identical autophagic patterns, and so did MDA231 breast cancer xenografts and HCT116 colon tumor spheroids (also analyzed by electron microscopy). Moreover, MCF-7 human breast cancer cell lines confirmed induction of LC3A by anoxia and Thapsigargin. It is concluded that autophagy can be readily recognized in breast carcinomas by light microscopy, after immunohistochemical staining with LC3A, but the significance of the various patterns expressed would need further evaluation.


Subject(s)
Autophagy , Breast Neoplasms/metabolism , Carcinoma/metabolism , Gene Expression Regulation, Neoplastic , Microtubule-Associated Proteins/biosynthesis , Adult , Aged , Breast Neoplasms/surgery , Carcinoma/surgery , Cell Line, Tumor , Female , Humans , Male , Microscopy, Electron/methods , Middle Aged , Neoplasm Transplantation , Treatment Outcome
6.
Ann N Y Acad Sci ; 1177: 185-97, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19845621

ABSTRACT

Eukaryotic chromatin is subject to multiple posttranslational histone modifications such as acetylation, methylation, phosphorylation, and ubiquitination. These various covalent modifications have been proposed to constitute a "histone code," playing important roles in the establishment of global chromatin environments, transcription, DNA repair, and DNA replication. Among these modifications, histone methylation specifies regulatory marks that delineate transcriptionally active and inactive chromatin. These histone methyl marks were considered irreversible; however, recent identification of site-specific histone demethylases demonstrates that histone methylation is dynamically regulated, which may allow cells to rapidly change chromatin conformation to adapt to environmental stresses or intrinsic stimuli. Of major interest is the observation that these histone demethylase enzymes, which are in the Jumonji gene family, require oxygen to function and, in some cases, are induced by hypoxia in an HIFalpha-dependent manner. This provides a new mechanism for regulation of the response to hypoxia.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Epigenesis, Genetic , Gene Expression Regulation , Oxidoreductases, N-Demethylating/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA-Binding Proteins , Humans , Jumonji Domain-Containing Histone Demethylases , Mice , Oxidoreductases, N-Demethylating/classification , Oxidoreductases, N-Demethylating/genetics , Phylogeny , Retinoblastoma-Binding Protein 2
7.
Prostate ; 69(14): 1507-20, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19544327

ABSTRACT

INTRODUCTION: New in vitro models of castration-resistant prostate cancer (CRPC) are urgently required. METHODS: Trans-rectal needle biopsies (TRBP) of the prostate were performed for research purposes on progressing CRPC patients who had not received prior treatment to the prostate. Biopsies were immediately digested with collagenase and plated onto collagen-coated flasks with a feeder layer of 3T6 cells and cultured in cytokine-supplemented keratinocyte serum-free medium. RESULTS: Biopsies from 25 patients were collected and one of these, following an initial period of crisis, spontaneously immortalized. A series of cell lines called Bob were then established from a clone that survived CD133-selection followed by 4 weeks under adhesion-independent conditions in methylcellulose. Gains and losses previously described in clinical prostate tumors, most notably loss of 8(p) and gain of 8(q), were identified on comparative genomic hybridization and long-term growth in culture, survival in methylcellulose and invasion through matrigel confirmed the malignant phenotype of Bob. Furthermore, Bob expressed high levels of p53 and markers of early differentiation, including K8, prostatic acid phosphatase and prostate stem cell antigen. There was, however, no in vivo growth and ERG and ETV1 were not rearranged. Growth in serum permitted some differentiation. CONCLUSION: This is the first spontaneously immortalized prostate cancer cell line to be established from a TRBP of a patient with CRPC. Bob is a novel pre-clinical model for functional studies in CRPC and especially for studying the CRPC "basal" phenotype.


Subject(s)
Cell Culture Techniques/methods , Cell Separation/methods , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Animals , Biopsy, Needle , Blood Proteins/pharmacology , Cell Adhesion , Cell Differentiation , Cell Division , Cell Line, Transformed , Cell Line, Tumor , Cell Movement , Comparative Genomic Hybridization , DNA, Neoplasm/analysis , Eukaryotic Initiation Factor-3/metabolism , Humans , Karyotyping , Male , Methylcellulose , Mice , Mice, SCID , Neoplasm Transplantation , Phenotype , Spheroids, Cellular
8.
Prostate ; 68(9): 1007-24, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18398820

ABSTRACT

BACKGROUND: Recent evidence suggests that prostate stem cells in benign and tumor tissue express the cell surface marker CD133, but these cells have not been well characterized. The aim of our study was to gene expression profile CD133-expressing cells. METHODS: We analyzed CD133-positive (CD133+) and -negative (CD133-) sub-populations of high-integrin expressing epithelial cells isolated from benign human prostate tissue and hormone-refractory prostate cancer (HRPC). RESULTS: CD133+ cells freshly isolated from benign prostate tissue exhibited an expression profile characteristic of a putative stem/progenitor cell population, with transcripts involved in biological processes ranging from development and ion homeostasis to cell communication. The profile of CD133- cells was consistent with that of a transit amplifying population, suggesting up-regulated proliferation and metabolism. Comparison of benign populations to those from HRPC showed some similarities between CD133+ profiles but also revealed significant differences that provide a tumor-specific pattern, which included evidence of increased metabolic activity and active proliferation. Subsequently, we demonstrated protein expression of a number of candidate genes in these cell populations and in benign tissue. In a novel observation we also found expression of some of these markers in prostate tumors, including the oligodendrocyte lineage transcription factor OLIG1. CONCLUSIONS: This study provides a unique genome-wide molecular signature of CD133+ and CD133- human prostate epithelial cells. This will provide a valuable resource for prostate stem cell biology research and the identification of novel therapeutic targets for the treatment of prostate cancer.


Subject(s)
Antigens, CD/biosynthesis , Glycoproteins/biosynthesis , Prostatic Hyperplasia/immunology , Prostatic Neoplasms/immunology , AC133 Antigen , Antigens, CD/genetics , Cluster Analysis , Epithelial Cells/immunology , Gene Expression Profiling , Glycoproteins/genetics , Humans , Male , Oligonucleotide Array Sequence Analysis , Peptides/genetics , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...