Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Front Oncol ; 11: 590771, 2021.
Article in English | MEDLINE | ID: mdl-33996536

ABSTRACT

BACKGROUND: Myoglobin (MB) is increasingly recognized as a key player in cancer growth and metastasis. Low oxygen tensions, commonly associated with highly aggressive and recurrent cancers, have been shown to regulate its expression in several cancers such as lung, neck, prostate and breast cancer. However, it is not yet known whether it contributes to the growth and spread of brain cancers especially Glioblastoma multiforme (GBM). METHODS: Here we investigate the expression of MB, and its correlation with the hypoxia markers carbonic anhydrase IX (CAIX) and lactate dehydrogenase A (LDHA), in human tissue microarrays of multiple organ tumors, brain tumors, and GBM tumors, and their respective cancer-adjacent normal tissues. Correlation between MB protein expression and tumor grade was also assessed. RESULTS: We show that MB protein is expressed in a wide variety of cancers, benign tumors, cancer-adjacent normal tissues, hyperplastic tissue samples and normal brain tissue, and low oxygen tensions modulate MB protein expression in different brain cancers, including GBM. Enhanced nuclear LDHA immune-reactivity in GBM was also observed. Finally, we report for the first time a positive correlation between MB expression and brain tumor grade. CONCLUSION: Our data suggest that hypoxia regulate MB expression in different brain cancers (including GBM) and that its expression is associated with a more aggressive phenotype as indicated by the positive correlation with the brain tumor grade. Additionally, a role for nuclear LDHA in promoting aggressive tumor phenotype is also suggested based on enhanced nuclear expression which was observed only in GBM.

2.
Oncol Rep ; 43(3): 975-985, 2020 03.
Article in English | MEDLINE | ID: mdl-32020230

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive human brain cancer. Little is known regarding how these cells adapt to the harsh tumor microenvironment, and consequently survive and resist various treatments. Myoglobin (MB), the oxygen­binding hemoprotein, has been shown to be ectopically expressed in different human cancers and cell lines, and its expression is hypothesized to be an adaptation mechanism to hypoxia. The aim of the present study was to determine whether cancer­related and hypoxia­responsive MB mRNA splice variants are expressed in human GBM cells and glioblastoma tumor xenografts, and whether their expression is induced by hypoxia and correlated with hypoxia markers [lactate dehydrogenase A (LDHA), glucose transporter 1 (GLUT1), vascular endothelial growth factor (VEGF) and carbonic anhydrase IX (CAIX)]. Conventional reverse transcription (RT)­PCR, DNA sequencing, RT­quantitative PCR and immunohistochemistry were conducted to investigate MB expression in hypoxia­sensitive (M010b, M059J) and ­tolerant (M059K, M006xLo) GBM cell lines that also exhibit differential response towards radiation, rendering them a valuable translational GBM model. It was revealed that cancer­related MB variants 9, 10, 11 and 13 were expressed in GBM cells under normoxia, and following hypoxia, their expression exhibited modest­to­significant upregulation that correlated with hypoxia markers. It was also demonstrated that MB was upregulated in hypoxic microregions of glioblastoma tumor xenografts that were stained in matched tumor regions of serial tumor sections with the hypoxia markers, pimonidazole, CAIX, VEGF and LDHA. The present study identified myoglobin as a potential contributor to the hypoxia adaptation and survival strategies of glioblastoma, and may explain the aggressiveness and frequent recurrence rates associated with GBM.


Subject(s)
Biomarkers, Tumor/genetics , Glioblastoma/genetics , Myoglobin/genetics , Tumor Hypoxia/genetics , Carbonic Anhydrase IX/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Glucose Transporter Type 1/genetics , Heterografts , Humans , L-Lactate Dehydrogenase/genetics , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Protein Isoforms/genetics , RNA, Messenger/genetics , Tumor Microenvironment/genetics , Vascular Endothelial Growth Factor A/genetics
3.
Oncol Rep ; 31(4): 1947-53, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24573635

ABSTRACT

Pathological angiogenesis is a characteristic feature of glioblastoma multiforme (GBM) where the balance between pro-angiogenic and anti-angiogenic factors are shifted towards the pro-angiogenic phenotype. In this study we sought to determine whether angiostatins are expressed by GBM cells and whether their expression along with other related factors [matrix metalloproteinase (MMP)-2, MMP-9, and collagen type I α1 (COLIA1)] are altered by hypoxia and/or correlated with the levels of cancer stem cell marker CD133. Using qRT-PCR, western blotting, and gelatin zymography, we examined the expression of angiostatins, MMP-2, MMP-9, COLIA1 and CD133 in GBM cell lines cultured under aerobic conditions and hypoxia. Expression levels of MMP-2 and MMP-9 were significantly induced by hypoxia. Angiostatins were detected in all GBM cell lines and were increased by hypoxia while the angiostatin isoform of 38-kDa was the most abundant in GBM cells under aerobic and hypoxic conditions. COLIA1 and CD133 were significantly increased in several GBM cell lines under hypoxia. Despite expression and upregulation of anti-angiogenic factors (e.g. angiostatins) in GBM cells, they are overwhelmed by the overexpression of a larger number of angiogenic factors that shift the angiogenic balance towards the pro-angiogenic phenotype. Thus, an exogenous administration of anti-angiogenic factors may be required to improve the treatment of GBM tumors.


Subject(s)
Angiostatins/biosynthesis , Glioblastoma/pathology , Neovascularization, Pathologic/metabolism , AC133 Antigen , Angiostatins/analysis , Antigens, CD/analysis , Antigens, CD/biosynthesis , Blotting, Western , Cell Hypoxia , Cell Line, Tumor , Collagen Type I/analysis , Collagen Type I/biosynthesis , Glioblastoma/metabolism , Glycoproteins/analysis , Glycoproteins/biosynthesis , Humans , Matrix Metalloproteinase 2/analysis , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/analysis , Matrix Metalloproteinase 9/biosynthesis , Neovascularization, Pathologic/pathology , Peptides/analysis , Reverse Transcriptase Polymerase Chain Reaction
4.
Int J Oncol ; 44(3): 950-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24379112

ABSTRACT

Hemoglobin is produced mainly in erythroid cells. However, it has been reported in non-erythroid cells of human and rodents. We have shown previously that neuroglobin, cytoglobin and hemoglobin are expressed in human glioblastoma multiforme (GBM) cells. We sought to determine whether hemoglobin expression is upregulated by hypoxia, and whether its expression is restricted to the cancer stem cell populations in different GBM cell lines or GBM brain tumor initiating cells (BTICs). Flow cytometry, magnetic cell sorting and qRT-PCR were used to examine the hypoxic upregulation of hemoglobins as well as erythropoietin (EPO) and erythropoietin receptor (EPOR) in GBM cell lines (M006x, M059J, M059K, U87R and U87T) and GBM-BTICs. The data showed significantly increased expression in globins (α, ß, γ, δ, ζ and ε), EPO and EPOR mRNA levels under hypoxia. Globin expression is not limited to the stem cell populations or GBM-BTICs but is a property of the entire GBM population. We assumed that the total expression of mRNA of different normalized globins (α, ß, γ, δ, ζ and ε) at different time­points for the same cell line is 100%. Under aerobic conditions, ε globin was predominantly expressed, and then decreased gradually with increasing time in hypoxia. This was coupled to a concomitant increase in α and γ globins. Our findings suggest that hypoxic upregulation of hemoglobin expression in GBM cells may be a part of a repertoire of active defence and adaptation mechanisms enabling these cells to acquire resistance to aggressive multimodality treatments of chemotherapy and radiotherapy. New therapeutic strategies to interfere with hemoglobin expression or function in GBM cells are required.


Subject(s)
Brain Neoplasms/genetics , Cell Hypoxia/genetics , Glioblastoma/genetics , Hemoglobins/biosynthesis , Brain Neoplasms/pathology , Erythropoietin/biosynthesis , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Hemoglobins/genetics , Humans , Neoplastic Stem Cells , Receptors, Erythropoietin/biosynthesis
5.
Int J Oncol ; 44(2): 514-20, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24270851

ABSTRACT

Hemoglobin is a hemoprotein, produced mainly in erythrocytes circulating in the blood. However, non-erythroid hemoglobins have been previously reported in other cell types including human and rodent neurons of embryonic and adult brain, but not astrocytes and oligodendrocytes. Human glioblastoma multiforme (GBM) is the most aggressive tumor among gliomas. However, despite extensive basic and clinical research studies on GBM cells, little is known about glial defence mechanisms that allow these cells to survive and resist various types of treatment. We have shown previously that the newest members of vertebrate globin family, neuroglobin (Ngb) and cytoglobin (Cygb), are expressed in human GBM cells. In this study, we sought to determine whether hemoglobin is also expressed in GBM cells. Conventional RT-PCR, DNA sequencing, western blot analysis, mass spectrometry and fluorescence microscopy were used to investigate globin expression in GBM cell lines (M006x, M059J, M059K, M010b, U87R and U87T) that have unique characteristics in terms of tumor invasion and response to radiotherapy and hypoxia. The data showed that α, ß, γ, δ, ζ and ε globins are expressed in all tested GBM cell lines. To our knowledge, we are the first to report expression of fetal, embryonic and adult hemoglobin in GBM cells under normal physiological conditions that may suggest an undefined function of those expressed hemoglobins. Together with our previous reports on globins (Ngb and Cygb) expression in GBM cells, the expression of different hemoglobins may constitute a part of series of active defence mechanisms supporting these cells to resist various types of treatments including chemotherapy and radiotherapy.


Subject(s)
Fetal Hemoglobin/metabolism , Glioblastoma/metabolism , Hemoglobins/metabolism , Adult , Fetal Hemoglobin/genetics , Fluorescent Antibody Technique , Glioblastoma/genetics , Hemoglobins/classification , Hemoglobins/genetics , Humans , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
6.
Breast Cancer Res ; 13(3): 210, 2011 May 31.
Article in English | MEDLINE | ID: mdl-21672271

ABSTRACT

Aberrant Notch signaling can induce mammary gland carcinoma in transgenic mice, and high expressions of Notch receptors and ligands have been linked to poor clinical outcomes in human patients with breast cancer. This suggests that inhibition of Notch signaling may be beneficial for breast cancer treatment. In this review, we critically evaluate the evidence that supports or challenges the hypothesis that inhibition of Notch signaling would be advantageous in breast cancer management. We find that there are many remaining uncertainties that must be addressed experimentally if we are to exploit inhibition of Notch signaling as a treatment approach in breast cancer. Nonetheless, Notch inhibition, in combination with other therapies, is a promising avenue for future management of breast cancer. Furthermore, since aberrant Notch4 activity can induce mammary gland carcinoma in the absence of RBPjκ, a better understanding of the components of RBPjκ-independent oncogenic Notch signaling pathways and their contribution to Notch-induced tumorigenesis would facilitate the deployment of Notch inhibition strategies for effective treatment of breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/metabolism , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/metabolism , Animals , Breast/metabolism , Breast/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic , Female , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/pathology , Mice , Mice, Transgenic , Molecular Targeted Therapy , Neoplastic Stem Cells/pathology , Receptors, Notch/genetics , Signal Transduction
7.
Radiat Res ; 176(2): 198-207, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21631290

ABSTRACT

Cytoglobin is a recently identified vertebrate globin whose functions include scavenging reactive oxygen and nitrosative species. In tumor cells, CYGB may function as a tumor suppressor gene. Here we show that knockdown of cytoglobin expression can sensitize human glioma cells to oxidative stress induced by chemical inhibitors of the electron transport chain and as well can increase cellular radiosensitivity. When treated with antimycin A, an inhibitor of the mitochondrial electron transport chain, cytoglobin-deficient cells showed significantly higher H2O2 levels, whereas H2O2 levels were significantly reduced in cytoglobin-overexpressing cells. In addition, cytoglobin knockdown significantly decreased the doubling time of glioma cell lines, consistent with a putative tumor suppressor function. These finding suggest that modulating cytoglobin levels may be a promising treatment strategy for sensitizing human glioma cells to oxidative stress that is induced by ionizing radiation, certain chemotherapies and ischemia-reperfusion.


Subject(s)
Gene Knockdown Techniques , Glioma/pathology , Globins/deficiency , Globins/genetics , Oxidative Stress/genetics , Oxidative Stress/radiation effects , Radiation Tolerance/genetics , Cell Cycle/genetics , Cell Cycle/radiation effects , Cell Hypoxia/genetics , Cell Hypoxia/radiation effects , Cell Line, Tumor , Cell Proliferation/radiation effects , Cell Survival/genetics , Cell Survival/radiation effects , Cytoglobin , Globins/metabolism , Humans , Reactive Oxygen Species/metabolism
8.
Biochem Biophys Res Commun ; 407(3): 479-85, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21414294

ABSTRACT

The Notch signaling pathway is essential for embryonic development, organogenesis, and tissue homeostasis. Aberrant Notch signaling is associated with several types of cancers. The active form of Notch receptor is its intracellular domain (NICD), which is released from the cell membrane by serial proteolytic cleavages following ligand binding. Dose-dependent effects of NICD on cellular phenotypes have been observed under several conditions although the underlying mechanisms have not been well studied. Moreover, there are four mammalian Notch paralogs that have redundant as well as unique functions. The molecular basis for this variability is also not well understood. In this study, we used size exclusion chromatography to examine the overall distribution of NICD among NICD-containing protein complexes under conditions of increasing NICD abundance. We found that the assembly of NICD protein complexes was dose-dependent and that the abundance of the canonical complex was limited by, MAML, one of the proteins involved in the formation of canonical NICD transactivation complex, which became saturated with increasing NICD abundance. In addition, N4ICD showed a unique elution profile among the four NICDs. These results help to explain the dose-dependent and paralog-specific activities of NICD. These results are informative for the development of new reagents to block Notch signaling for therapeutic benefit.


Subject(s)
DNA-Binding Proteins/chemistry , Multiprotein Complexes/chemistry , Receptors, Notch/chemistry , Transcription Factors/chemistry , Cell Line, Tumor , Humans , Protein Structure, Tertiary , Receptors, Notch/genetics
9.
Med Phys ; 37(7): 3855-61, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20831093

ABSTRACT

PURPOSE: Magnetic resonance imaging was used to compare the responses of human glioma tumor xenografts to a single fraction of radiation, where a change in radiosensitivity was induced by use of a suture-based ligature. METHODS: Ischemia was induced by use of a suture-based ligature. Six mice were treated with 800 cGy of 200 kVp x rays while the ligature was applied. An additional six mice had the ligature applied for the same length of time but were not irradiated. Quantitative maps of each tumor were produced of water apparent diffusion coefficient (ADC) and transverse relaxation time (T2). Mice were imaged before and at multiple points after treatment. Volumetric, ADC, and T2 responses of the ligated groups were compared to previously measured responses of the same tumor model to the same radiation treatment, as well as those from an untreated control group. RESULTS: Application of the ligature without irradiation did not affect tumor ADC values, but did produce a temporary decrease in tumor T2 values. Average tumor T2 was reduced by 6.2% 24 h after the ligature was applied. Average tumor ADC increased by 9.6% 7 days after irradiation with a ligature applied. This response was significantly less than that observed in the same tumor model when no ligature is present (21.8% at 7 days after irradiation). CONCLUSIONS: These observations indicate that the response of ADC to radiation therapy is not determined entirely by physical dose deposition, but at least in part by radiosensitivity and resultant biological response.


Subject(s)
Radiation Tolerance/radiation effects , Radiotherapy , Animals , Cell Line, Tumor , Diffusion , Humans , Mice , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/physiopathology , Neoplasms/radiotherapy , Oxygen/metabolism , Radiotherapy Dosage , Time Factors
10.
Cancer Cell Int ; 10: 33, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20828399

ABSTRACT

BACKGROUND: Cytoglobin (Cygb) and neuroglobin (Ngb) are recently identified globin molecules that are expressed in vertebrate tissues. Upregulation of Cygb and Ngb under hypoxic and/or ischemic conditions in vitro and in vivo increases cell survival, suggesting possible protective roles through prevention of oxidative damage. We have previously shown that Ngb is expressed in human glioblastoma multiforme (GBM) cell lines, and that expression of its transcript and protein can be significantly increased after exposure to physiologically relevant levels of hypoxia. In this study, we extended this work to determine whether Cygb is also expressed in GBM cells, and whether its expression is enhanced under hypoxic conditions. We also compared Cygb and Ngb expression in human primary tumor specimens, including brain tumors, as well as in human normal tissues. Immunoreactivity of carbonic anhydrase IX (CA IX), a hypoxia-inducible metalloenzyme that catalyzes the hydration of CO2 to bicarbonate, was used as an endogenous marker of hypoxia. RESULTS: Cygb transcript and protein were expressed in human GBM cells, and this expression was significantly increased in most cells following 48 h incubation under hypoxia. We also showed that Cygb and Ngb are expressed in both normal tissues and human primary cancers, including GBM. Among normal tissues, Cygb and Ngb expression was restricted to distinct cell types and was especially prominent in ductal cells. Additionally, certain normal organs (e.g. stomach fundus, small bowel) showed distinct regional co-localization of Ngb, Cygb and CA IX. In most tumors, Ngb immunoreactivity was significantly greater than that of Cygb. In keeping with previous in vitro results, tumor regions that were positively stained for CA IX were also positive for Ngb and Cygb, suggesting that hypoxic upregulation of Ngb and Cygb also occurs in vivo. CONCLUSIONS: Our finding of hypoxic up-regulation of Cygb/Ngb in GBM cell lines and human tumor tissues suggests that these globin molecules may be part of the repertoire of defense mechanisms that allow cancer cells to survive in hypoxic microenvironments.

11.
Phys Med Biol ; 55(5): 1381-93, 2010 Mar 07.
Article in English | MEDLINE | ID: mdl-20150684

ABSTRACT

The purpose of this study is to investigate the response of transverse relaxation time (T2) and apparent diffusion coefficient (ADC) in human glioma tumor xenografts during and after fractionated radiotherapy. Tumor-bearing mice were divided into four treatment groups (n=6 per group) that received a total dose of 800 cGy of 200 kVp x-rays, given over two or three fractions, with a fraction spacing of either 24 or 72 h. A fifth treatment group received 800 cGy in a single fraction, and a sixth group of mice served as an untreated control. All mice were scanned pretreatment, before each fraction and at multiple points after treatment using a 9.4 T magnetic resonance imaging (MRI) system. Quantitative T2 and ADC maps were produced. All treated groups showed an increase in mean tumor ADC, though the time for this response to reach a maximum and return toward baseline was delayed in the fractionated groups. The highest ADC was measured 7 days after the final fraction of treatment for all groups. There were no significant differences in the maximum measured change in ADC between any of the treated groups, with the average measured maximum value being 20.5% above baseline. After treatment, all groups showed an increase in mean tumor T2, with the average measured maximum T2 being 4.7% above baseline. This increase was followed by a transition to mean T2 values below baseline values, with the average measured tumor T2 being 92.4% of the pretreatment value. The transition between elevated and depressed T2 values was delayed in the cases of fractionated therapies and occurred between 3.6 and 7.3 days after the last fraction of treatment. These results further the understanding of the temporal evolution of T2 and ADC during fractionated radiotherapy and support their potential use as time-sensitive biomarkers for tumor response.


Subject(s)
Diffusion , Dose Fractionation, Radiation , Radiotherapy/methods , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Disease Models, Animal , Humans , Mice , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/radiotherapy , Radiotherapy Dosage , Time Factors
12.
Med Phys ; 36(7): 2948-54, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19673193

ABSTRACT

The purpose of this study is to use magnetic resonance imaging to monitor the response of human glioma tumor xenografts to single fraction radiation therapy. Mice were divided into four treatment groups (n = 6 per group) that received 50, 200, 400, or 800 cGy of 200 kVp x rays. A fifth group (n = 6) received no radiation dose and served as the control. Quantitative maps of the treated tumor tissue were produced of water apparent diffusion coefficient (ADC) and transverse relaxation time (T2). Mice were imaged before and at multiple time points after treatment. There was a statistically significant difference in tumor growth relative to that of the control for all treatment groups. Only the highest dose group showed T2 values that were significantly different at all measured time points after treatment. In this group, there was an 8.3% increase in T2 relative to controls 2 days after treatment, but when measured 14 days after treatment, mean tumor T2 had dropped to 10.1% below the initial value. ADC showed statistically significant differences from the control at all dose points. A radiation dose dependence was observed. In the highest dose group, the fractional increases in ADC were higher than those observed for T2. ADC was sensitive to radiation-induced changes in lower dose groups that did not have significant T2 change. At all doses, elevation of mean tumor ADC preceded deviations in tumor growth from the control. These observations support the potential application of ADC as a time and dose sensitive marker of tumor response to radiation therapy.


Subject(s)
Dose Fractionation, Radiation , Glioblastoma/radiotherapy , Animals , Disease Models, Animal , Dose-Response Relationship, Radiation , Glioblastoma/pathology , Humans , Image Interpretation, Computer-Assisted , Magnetic Resonance Imaging/methods , Mice , Mice, Nude , Neoplasm Transplantation , Time Factors , Transplantation, Heterologous , Treatment Outcome
13.
Breast Cancer Res ; 11(4): R57, 2009.
Article in English | MEDLINE | ID: mdl-19660128

ABSTRACT

INTRODUCTION: Notch is a family of transmembrane protein receptors whose activation requires proteolytic cleavage by gamma-secretase. Since aberrant Notch signaling can induce mammary carcinomas in transgenic mice and high expression levels of Notch receptors and ligands correlates with overall poor clinical outcomes, inhibiting gamma-secretase with small molecules may be a promising approach for breast cancer treatment. Consistent with this hypothesis, two recent papers reported that gamma-secretase inhibitor I (GSI I), Z-LLNle-CHO, is toxic to breast cancer cells both in vitro and in vivo. In this study, we compared the activity and cytotoxicity of Z-LLNle-CHO to that of two highly specific GSIs, DAPT and L-685,458 and three structurally unrelated proteasome inhibitors, MG132, lactacystin, and bortezomib in order to study the mechanism underlying the cytotoxicity of Z-LLNle-CHO in breast cancer cells. METHODS: Three estrogen receptor (ER) positive cell lines, MCF-7, BT474, and T47D, and three ER negative cell lines, SKBR3, MDA-MB-231, and MDA-MB-468, were used in this study. Both SKBR3 and BT474 cells also overexpress HER2/neu. Cytotoxicity was measured by using an MTS cell viability/proliferation assay. Inhibition of gamma-secretase activity was measured by both immunoblotting and immunofluorescent microscopy in order to detect active Notch1 intracellular domain. Proteasome inhibition was determined by using a cell-based proteasome activity assay kit, by immunoblotting to detect accumulation of polyubiquitylated protein, and by immunofluorescent microscopy to detect redistribution of cellular ubiquitin. RESULTS: We found that blocking gamma-secretase activity by DAPT and L-685,458 had no effect on the survival and proliferation of a panel of six breast cancer cell lines while Z-LLNle-CHO could cause cell death even at concentrations that inhibited gamma-secretase activity less efficiently. Furthermore, we observed that Z-LLNle-CHO could inhibit proteasome activity and the relative cellular sensitivity of these six breast cancer cell lines to Z-LLNle-CHO was the same as observed for three proteasome inhibitors. Finally, we found that the cell killing effect of Z-LLNle-CHO could be reversed by a chemical that restored the proteasome activity. CONCLUSIONS: We conclude that the cytotoxicity of Z-LLNle-CHO in breast cancer cells is mediated by proteasome inhibition, not by gamma-secretase inhibition.


Subject(s)
Adenocarcinoma/pathology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Breast Neoplasms/pathology , Neoplasm Proteins/antagonists & inhibitors , Oligopeptides/toxicity , Proteasome Inhibitors , Adenocarcinoma/enzymology , Breast Neoplasms/enzymology , Carbamates/pharmacology , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , Cell Line, Tumor/pathology , Dipeptides/pharmacology , Drug Delivery Systems , Estrogens , Female , Humans , Neoplasm Proteins/analysis , Neoplasm Proteins/metabolism , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/pathology , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/drug effects , Receptor, Notch1/metabolism , Receptors, Estrogen/analysis
14.
Mol Oncol ; 3(1): 45-53, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19383366

ABSTRACT

Neuroglobin is a recently identified globin molecule that is expressed predominantly in the vertebrate brain. Neuroglobin expression increases in oxygen-deprived neurons, suggesting it protects neurons from ischemic cell death. We report that neuroglobin transcript and protein are expressed in human glioblastoma cells, and that this expression increases in hypoxia in vitro. We also show that neuroglobin is up-regulated in hypoxic microregions of glioblastoma tumor xenografts. Our finding of hypoxic up-regulation of neuroglobin in human glioblastoma cells may provide insight into how tumor cells adapt to and survive in hypoxic microenvironments.


Subject(s)
Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Globins/genetics , Hypoxia/genetics , Nerve Tissue Proteins/genetics , Animals , Cell Line, Tumor , Glioblastoma/genetics , Humans , Mice , Neoplasms, Experimental , Neuroglobin , Transplantation, Heterologous , Up-Regulation/genetics
15.
Cancer Res ; 67(14): 6619-25, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17638872

ABSTRACT

Human polynucleotide kinase (hPNK) is a bifunctional enzyme possessing a 5'-DNA kinase activity and a 3'-phosphatase activity. Studies based on cell extracts and purified proteins have indicated that hPNK can act on single-strand breaks and double-strand breaks (DSB) to restore the termini to the chemical form required for further action by DNA repair polymerases and ligases (i.e., 5'-phosphate and 3'-hydroxyl termini). These studies have revealed that hPNK can bind to XRCC4, and as a result, hPNK has been implicated as a participant in the nonhomologous end joining (NHEJ) pathway for DSB repair. We sought to confirm the role of hPNK in NHEJ in the cellular setting using a genetic approach. hPNK was stably down-regulated by RNA interference expression in M059K glioblastoma cells, which are NHEJ positive, and M059J cells, which are NHEJ deficient due to a lack of DNA-PK catalytic subunit (DNA-PKcs). Whereas depletion of hPNK significantly sensitized M059K cells to ionizing radiation, no additional sensitization was conferred to M059J cells, clearly implying that hPNK operates in the same DNA repair pathway as DNA-PKcs. On the other hand, depletion of hPNK did not increase the level of sister chromatid exchanges, indicating that hPNK is not involved in the homologous recombination DSB repair pathway. We also provide evidence that the action of hPNK in the repair of camptothecin-induced topoisomerase 1 "dead-end" complexes is independent of DNA-PKcs and that hPNK is not involved in the nucleotide excision repair pathway.


Subject(s)
DNA Breaks, Double-Stranded , DNA Damage , Polynucleotide 5'-Hydroxyl-Kinase/genetics , Polynucleotide 5'-Hydroxyl-Kinase/physiology , Antineoplastic Agents, Phytogenic/pharmacology , Brain Neoplasms/metabolism , Camptothecin/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , DNA Repair , Dose-Response Relationship, Drug , Glioblastoma/metabolism , Histones/metabolism , Humans , Recombination, Genetic , Sister Chromatid Exchange
16.
Cancer Cell ; 11(1): 37-51, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17222789

ABSTRACT

The unique metabolic profile of cancer (aerobic glycolysis) might confer apoptosis resistance and be therapeutically targeted. Compared to normal cells, several human cancers have high mitochondrial membrane potential (DeltaPsim) and low expression of the K+ channel Kv1.5, both contributing to apoptosis resistance. Dichloroacetate (DCA) inhibits mitochondrial pyruvate dehydrogenase kinase (PDK), shifts metabolism from glycolysis to glucose oxidation, decreases DeltaPsim, increases mitochondrial H2O2, and activates Kv channels in all cancer, but not normal, cells; DCA upregulates Kv1.5 by an NFAT1-dependent mechanism. DCA induces apoptosis, decreases proliferation, and inhibits tumor growth, without apparent toxicity. Molecular inhibition of PDK2 by siRNA mimics DCA. The mitochondria-NFAT-Kv axis and PDK are important therapeutic targets in cancer; the orally available DCA is a promising selective anticancer agent.


Subject(s)
Apoptosis/physiology , Mitochondria/metabolism , Neoplasms/metabolism , Potassium Channels/metabolism , Animals , Cell Line, Tumor , Dichloroacetic Acid/pharmacology , Humans , Immunoblotting , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Microscopy, Confocal , Mitochondria/drug effects , NFATC Transcription Factors/metabolism , Patch-Clamp Techniques , Potassium Channels/drug effects , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Rats , Rats, Nude , Reverse Transcriptase Polymerase Chain Reaction
17.
Radiat Res ; 165(3): 283-92, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16494516

ABSTRACT

Rapid phosphorylation of histone H2AX after exposure of cells to ionizing radiation occurs at DSB sites and extends to a region including as much as 30 Mbp of chromatin to form visible microscopic structures called gamma-H2AX foci. Although the kinetics of total cellular histone H2AX phosphorylation after irradiation has been characterized, we still know little about the phosphorylation kinetics of individual gamma-H2AX foci. In addition, there are hundreds of smaller gamma-H2AX foci that are not associated with DNA double-strand breaks. We refer to these sites as DSB-unrelated gamma-H2AX foci. By using indirect immunofluorescence microscopy, deconvolution and three-dimensional image analysis, we established an objective method to quantitatively analyze each gamma-H2AX focus as well as to discriminate DSB-related gamma-H2AX foci from DSB-unrelated gamma-H2AX foci. Using this method, we found that histone H2AX phosphorylation at different DSB sites was asynchronous after exposure to ionizing radiation. This may reflect the heterogeneous characteristic of free DNA ends that are generated under these conditions. In addition, we found that increased histone H2AX phosphorylation also occurred outside of DSB sites after exposure to ionizing radiation. The function of this DSB-unassociated phosphorylation is not known.


Subject(s)
DNA Damage/genetics , DNA/genetics , Histones/metabolism , Cell Line, Tumor , Histones/genetics , Humans , Kinetics , Phosphorylation/radiation effects , Radiation, Ionizing , S Phase/genetics , S Phase/radiation effects , Time Factors
18.
Cancer Lett ; 243(2): 193-201, 2006 Nov 18.
Article in English | MEDLINE | ID: mdl-16412569

ABSTRACT

We tested whether mtDNA mutations are associated with poor outcome in patients with invasive cervix cancer. Tumor samples were banked more than 10 years ago from women with diagnoses of invasive cervix cancer. Automated techniques were used to determine the sequence of the mtDNA-encoded Complex I subunits. Approximately one-third of all tumors had multiple mtDNA sequence alterations. Both univariate and multivariate analysis of the 10 years survival probability showed that the 10 years survival of patients whose tumors had eight or more nucleotide substitutions was significantly worse (P<0.0063 and P<0.012, respectively). The log-rank test also found a significant difference in overall survival (P<0.003). These results suggest that multiple mtDNA mutations are an independent marker of poor prognosis, and that prospective clinical trials that incorporate analysis of mitochondrial genetic alterations in cervix cancer are warranted.


Subject(s)
DNA, Mitochondrial/genetics , Mutation/genetics , Uterine Cervical Neoplasms/pathology , Adult , DNA Mutational Analysis , DNA, Mitochondrial/chemistry , Electron Transport Complex I/genetics , Female , Humans , Middle Aged , Multivariate Analysis , Neoplasm Invasiveness , Prognosis , Retrospective Studies , Survival Analysis , Survival Rate , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/mortality
19.
Mol Cancer ; 3: 19, 2004 Jul 12.
Article in English | MEDLINE | ID: mdl-15248896

ABSTRACT

BACKGROUND: Hypoxia-tolerant human glioma cells reduce oxygen consumption rate in response to oxygen deficit, a defense mechanism that contributes to survival under moderately hypoxic conditions. In contrast, hypoxia-sensitive cells lack this ability. As it has been previously shown that hypoxia-tolerant (M006x, M006xLo, M059K) and -sensitive (M010b) glioma cells express differences in mitochondrial function, we investigated whether mitochondrial DNA-encoded mutations are associated with differences in the initial response to oxygen deficit. RESULTS: The mitochondrial genome was sequenced and 23 mtDNA alterations were identified, one of which was an unreported mutation (T-C transition in base pair 14634) in the hypoxia-sensitive cell line, M010b, that resulted in a single amino acid change in the gene encoding the ND6 subunit of NADH:ubiquinone oxidoreductase (Complex I). The T14634C mutation did not abrogate ND6 protein expression, however, M010b cells were more resistant to rotenone, an agent used to screen for Complex I mutations, and adriamycin, an agent activated by redox cycling. The specific function of mtDNA-encoded, membrane-embedded Complex I ND subunits is not known at present. Current models suggest that the transmembrane arm of Complex I may serve as a conformationally driven proton channel. As cellular respiration is regulated, in part, by proton flux, we used homology-based modeling and computational molecular biology to predict the 3D structure of the wild type and mutated ND6 proteins. These models predict that the T14634C mutation alters the structure and orientation of the trans-membrane helices of the ND6 protein. CONCLUSION: Complex I ND subunits are mutational hot spots in tumor mtDNA. Genetic changes that alter Complex I structure and function may alter a cell's ability to respond to oxygen deficit and consolidate hypoxia rescue mechanisms, and may contribute to resistance to chemotherapeutic agents that require redox cycling for activation.


Subject(s)
Central Nervous System Neoplasms/genetics , DNA, Mitochondrial/genetics , Electron Transport Complex I/genetics , Glioma/genetics , Hypoxia/genetics , Mutation/genetics , Cell Growth Processes/genetics , Cell Line, Tumor , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/pathology , DNA Mutational Analysis/methods , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/genetics , Energy Metabolism/genetics , Glioma/metabolism , Glioma/pathology , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Nuclear Proteins/genetics , Oxidative Phosphorylation , Protein Biosynthesis/genetics , Protein Subunits/genetics , Sequence Homology, Nucleic Acid , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...