Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33649219

ABSTRACT

Infiltration of tumor-promoting immune cells is a strong driver of tumor progression. Especially the accumulation of macrophages in the tumor microenvironment is known to facilitate tumor growth and to correlate with poor prognosis in many tumor types. TAp73, a member of the p53/p63/p73 family, acts as a tumor suppressor and has been shown to suppress tumor angiogenesis. However, what role TAp73 has in regulating immune cell infiltration is unknown. Here, we report that low levels of TAp73 correlate with an increased NF-κB-regulated inflammatory signature in breast cancer. Furthermore, we show that loss of TAp73 results in NF-κB hyperactivation and secretion of Ccl2, a known NF-κB target and chemoattractant for monocytes and macrophages. Importantly, TAp73-deficient tumors display an increased accumulation of protumoral macrophages that express the mannose receptor (CD206) and scavenger receptor A (CD204) compared to controls. The relevance of TAp73 expression in human breast carcinoma was further accentuated by revealing that TAp73 expression correlates negatively with the accumulation of protumoral CD163+ macrophages in breast cancer patient samples. Taken together, our findings suggest that TAp73 regulates macrophage accumulation and phenotype in breast cancer through inhibition of the NF-κB pathway.


Subject(s)
Breast Neoplasms/immunology , NF-kappa B/immunology , Signal Transduction/immunology , Tumor Microenvironment/immunology , Tumor Protein p73/immunology , Tumor-Associated Macrophages/immunology , Animals , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Breast Neoplasms/pathology , Chemokine CCL2/immunology , Female , Humans , Membrane Glycoproteins/immunology , Mice , Receptors, Cell Surface/immunology , Receptors, Immunologic/immunology , Scavenger Receptors, Class A/immunology , Tumor-Associated Macrophages/pathology
2.
Proc Natl Acad Sci U S A ; 117(33): 20127-20138, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32747535

ABSTRACT

Medulloblastoma is the most common malignant brain tumor in children. Here we describe a medulloblastoma model using Induced pluripotent stem (iPS) cell-derived human neuroepithelial stem (NES) cells generated from a Gorlin syndrome patient carrying a germline mutation in the sonic hedgehog (SHH) receptor PTCH1. We found that Gorlin NES cells formed tumors in mouse cerebellum mimicking human medulloblastoma. Retransplantation of tumor-isolated NES (tNES) cells resulted in accelerated tumor formation, cells with reduced growth factor dependency, enhanced neurosphere formation in vitro, and increased sensitivity to Vismodegib. Using our model, we identified LGALS1 to be a GLI target gene that is up-regulated in both Gorlin tNES cells and SHH-subgroup of medulloblastoma patients. Taken together, we demonstrate that NES cells derived from Gorlin patients can be used as a resource to model medulloblastoma initiation and progression and to identify putative targets.


Subject(s)
Hedgehog Proteins/metabolism , Medulloblastoma/genetics , Neural Stem Cells/physiology , Anilides/pharmacology , Animals , Basal Cell Nevus Syndrome/genetics , Basal Cell Nevus Syndrome/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Galectin 1/genetics , Galectin 1/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Hedgehog Proteins/genetics , Humans , Mice , Neoplasms, Experimental , Patched-1 Receptor/genetics , Pyridines/pharmacology
3.
Oncogene ; 37(27): 3729-3739, 2018 07.
Article in English | MEDLINE | ID: mdl-29628507

ABSTRACT

Cellular responses to low oxygen conditions are mainly regulated by the Hypoxia-inducible factors (HIFs). Induction of HIF-1α in tumor cells activates the angiogenic switch and allows for metabolic adaptations. HIF-1α protein levels are tightly regulated through ubiquitin-mediated proteosomal degradation; however, high levels of HIF-1α is a common feature in many solid tumors and is thought to enhance cancer cell proliferation, migration, and survival. Here, we report that the oncogenic p73 isoform, ∆Np73, increases HIF-1α protein stability. We found that ∆Np73 represses expression of genes encoding subunits of the ECV complex, in particular Elongin C, Elongin B, Cullin 2, and Rbx1. The ECV complex is an E3 ligase complex responsible for polyubiquitinating HIF-1α. Loss of ∆Np73 increases ubiquitination of HIF-1α, leading to its degradation via the proteosomal pathway, and subsequent decrease of HIF-1α target genes. Taken together, our data demonstrates that high levels of ∆Np73 stabilize HIF-1α protein, allowing for it to accumulate and further potentiating its transcriptional activity and supporting tumor progression.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Tumor Protein p73/genetics , Tumor Protein p73/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Carrier Proteins/biosynthesis , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Cullin Proteins/biosynthesis , Elongin/biosynthesis , Humans , MCF-7 Cells , Mice , Mice, Nude , RNA Interference , RNA, Small Interfering/genetics , Ubiquitination/genetics
4.
Cell Oncol (Dordr) ; 40(6): 631-638, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28677036

ABSTRACT

PURPOSE: Multidrug resistance (MDR) is a major cause of treatment failure. In cancer cells, MDR is often caused by an increased efflux of therapeutic drugs mediated by an up-regulation of ATP binding cassette (ABC) transporters. It has previously been shown that oncogenic ΔNp73 plays an important role in chemo-resistance. Here we aimed at unraveling the role of ΔNp73 in regulating multidrug resistance in breast cancer and melanoma cells. METHODS: KEGG pathway analysis was used to identify pathways enriched in breast cancer samples with a high ΔNp73 expression. We found that the ABC transporter pathway was most enriched. The expression of selected ABC transporters was analyzed using qRT-PCR upon siRNA/shRNA-mediated knockdown or exogenous overexpression of ΔNp73 in the breast cancer-derived cell lines MCF7 and MDA-MB-231, as well as in primary melanoma samples and in the melanoma-derived cell line SK-MEL-28. The ability to efflux doxorubicin and the concomitant effects on cell proliferation were assessed using flow cytometry and WST-1 assays. RESULTS: We found that high ΔNp73 levels correlate with a general up-regulation of ABC transporters in breast cancer samples. In addition, we found that exogenous expression of ΔNp73 led to an increase in the expression of ABCB1 and ABCB5 in the breast cancer-derived cell lines tested, while knocking down of ΔNp73 resulted in a reduction in ABCB1 and ABCB5 expression. In addition, we found that ΔNp73 reduction leads to an intracellular retention of doxorubicin in MDA-MB-231 and MCF7 cells and a concomitant decrease in cell proliferation. The effect of ΔNp73 on ABCB5 expression was further confirmed in metastases from melanoma patients and in the melanoma-derived cell line SK-MEL-28. CONCLUSIONS: Our data support a role for ΔNp73 in the multidrug-resistance of breast cancer and melanoma cells.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Breast Neoplasms/metabolism , Melanoma/metabolism , Tumor Protein p73/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Blotting, Western , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , MCF-7 Cells , Melanoma/genetics , Real-Time Polymerase Chain Reaction , Tumor Protein p73/genetics
6.
PLoS One ; 10(5): e0119549, 2015.
Article in English | MEDLINE | ID: mdl-25932953

ABSTRACT

Malignant mesothelioma (MM) is an aggressive type of tumour causing high mortality. One reason for this paradigm may be the existence of a subpopulation of tumour-initiating cells (TICs) that endow MM with drug resistance and recurrence. The objective of this study was to identify and characterise a TIC subpopulation in MM cells, using spheroid cultures, mesospheres, as a model of MM TICs. Mesospheres, typified by the stemness markers CD24, ABCG2 and OCT4, initiated tumours in immunodeficient mice more efficiently than adherent cells. CD24 knock-down cells lost the sphere-forming capacity and featured lower tumorigenicity. Upon serial transplantation, mesospheres were gradually more efficiently tumrigenic with increased level of stem cell markers. We also show that mesospheres feature mitochondrial and metabolic properties similar to those of normal and cancer stem cells. Finally, we show that mesothelioma-initiating cells are highly susceptible to mitochondrially targeted vitamin E succinate. This study documents that mesospheres can be used as a plausible model of mesothelioma-initiating cells and that they can be utilised in the search for efficient agents against MM.


Subject(s)
Antineoplastic Agents/pharmacology , Lung Neoplasms/pathology , Mesothelioma/pathology , Neoplastic Stem Cells/pathology , Animals , Biomarkers, Tumor/metabolism , CD24 Antigen/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Gene Knockdown Techniques , Humans , Inhibitory Concentration 50 , Lung Neoplasms/metabolism , Mesothelioma/metabolism , Mesothelioma, Malignant , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasm Invasiveness , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Phenotype , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Tocopherols/pharmacology
7.
BMC Cancer ; 15: 401, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25967547

ABSTRACT

BACKGROUND: Accumulating evidence suggests that breast cancer involves tumour-initiating cells (TICs), which play a role in initiation, metastasis, therapeutic resistance and relapse of the disease. Emerging drugs that target TICs are becoming a focus of contemporary research. Mitocans, a group of compounds that induce apoptosis of cancer cells by destabilising their mitochondria, are showing their potential in killing TICs. In this project, we investigated mitochondrially targeted vitamin E succinate (MitoVES), a recently developed mitocan, for its in vitro and in vivo efficacy against TICs. METHODS: The mammosphere model of breast TICs was established by culturing murine NeuTL and human MCF7 cells as spheres. This model was verified by stem cell marker expression, tumour initiation capacity and chemotherapeutic resistance. Cell susceptibility to MitoVES was assessed and the cell death pathway investigated. In vivo efficacy was studied by grafting NeuTL TICs to form syngeneic tumours. RESULTS: Mammospheres derived from NeuTL and MCF7 breast cancer cells were enriched in the level of stemness, and the sphere cells featured altered mitochondrial function. Sphere cultures were resistant to several established anti-cancer agents while they were susceptible to MitoVES. Killing of mammospheres was suppressed when the mitochondrial complex II, the molecular target of MitoVES, was knocked down. Importantly, MitoVES inhibited progression of syngeneic HER2(high) tumours derived from breast TICs by inducing apoptosis in tumour cells. CONCLUSIONS: These results demonstrate that using mammospheres, a plausible model for studying TICs, drugs that target mitochondria efficiently kill breast tumour-initiating cells.


Subject(s)
Breast Neoplasms/metabolism , Electron Transport Complex II/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Tocopherols/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm , Female , Humans , MCF-7 Cells , Mice , Mice, Transgenic , Spheroids, Cellular , Tumor Burden/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Proc Natl Acad Sci U S A ; 112(1): 220-5, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25535357

ABSTRACT

The p53-family member TAp73 is known to function as a tumor suppressor and regulates genomic integrity, cellular proliferation, and apoptosis; however, its role in tumor angiogenesis is poorly understood. Here we demonstrate that TAp73 regulates tumor angiogenesis through repression of proangiogenic and proinflammatory cytokines. Importantly, loss of TAp73 results in highly vascularized tumors, as well as an increase in vessel permeability resulting from disruption of vascular endothelial-cadherin junctions between endothelial cells. In contrast, loss of the oncogenic p73 isoform ΔNp73 leads to reduced blood vessel formation in tumors. Furthermore, we show that up-regulated ΔNp73 levels are associated with increased angiogenesis in human breast cancer and that inhibition of TAp73 results in an accumulation of HIF-1α and up-regulation of HIF-1α target genes. Taken together, our data demonstrate that loss of TAp73 or ΔNp73 up-regulation activates the angiogenic switch that stimulates tumor growth and progression.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neovascularization, Pathologic/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Breast Neoplasms/pathology , Cadherins/metabolism , Cell Hypoxia , Cell Line, Transformed , Cell Proliferation , Endothelial Cells/metabolism , Female , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/pathology , Mice , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic , Permeability , Protein Isoforms/metabolism , Tumor Protein p73 , Zebrafish
9.
Free Radic Biol Med ; 67: 41-50, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24145120

ABSTRACT

Tumor-initiating cells (TICs) often survive therapy and give rise to second-line tumors. We tested the plausibility of sphere cultures as models of TICs. Microarray data and microRNA data analysis confirmed the validity of spheres as models of TICs for breast and prostate cancer as well as mesothelioma cell lines. Microarray data analysis revealed the Trp pathway as the only pathway upregulated significantly in all types of studied TICs, with increased levels of indoleamine-2,3-dioxygenase-1 (IDO1), the rate-limiting enzyme of Trp metabolism along the kynurenine pathway. All types of TICs also expressed higher levels of the Trp uptake system consisting of CD98 and LAT1 with functional consequences. IDO1 expression was regulated via both transcriptional and posttranscriptional mechanisms, depending on the cancer type. Serial transplantation of TICs in mice resulted in gradually increased IDO1. Mitocans, represented by α-tocopheryl succinate and mitochondrially targeted vitamin E succinate (MitoVES), suppressed IDO1 in TICs. MitoVES suppressed IDO1 in TICs with functional mitochondrial complex II, involving transcriptional and posttranscriptional mechanisms. IDO1 increase and its suppression by VE analogues were replicated in TICs from primary human glioblastomas. Our work indicates that IDO1 is increased in TICs and that mitocans suppress the protein.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Gene Expression Regulation, Neoplastic , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Mitochondria/drug effects , Neoplastic Stem Cells/drug effects , alpha-Tocopherol/pharmacology , Cell Line, Tumor , Electron Transport Complex II/genetics , Electron Transport Complex II/metabolism , Female , Fusion Regulatory Protein-1/genetics , Fusion Regulatory Protein-1/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kynurenine/metabolism , Large Neutral Amino Acid-Transporter 1/genetics , Large Neutral Amino Acid-Transporter 1/metabolism , Male , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Mitochondria/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction , Tryptophan/metabolism
11.
Antioxid Redox Signal ; 15(12): 2989-3002, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21504364

ABSTRACT

SIGNIFICANCE: Recent research has shown that tumors contain a small subpopulation of stem-like cells that are more resistant to therapy and that are likely to produce second-line tumors. RECENT ADVANCES: Cancer stem-like cells (CSCs) have been characterized by a variety of markers, including, for a number of types of cancer, high expression of the plasma membrane protein CD133, which is also indicative of the increase of stemness of cultured cancer cells growing as spheres. CRITICAL ISSUES: While the function of this protein has not yet been clearly defined, it may have a role in the stem-like phenotype of CSCs that cause (re-)initiation of tumors as well as their propagation. We hypothesize that CD133 selects for CSC survival against not only immunosurveillance mechanisms but also stress-induced apoptosis. FUTURE DIRECTIONS: High level of expression of CD133 may be a useful marker of more aggressive tumors that are recalcitrant toward established therapies. Compelling preliminary data indicate that drugs targeting mitochondria may be utilized as a novel, efficient cancer therapeutic modality.


Subject(s)
Antigens, CD/metabolism , Apoptosis , Glycoproteins/metabolism , Immunologic Surveillance , Mitochondria/metabolism , Peptides/metabolism , AC133 Antigen , Animals , Antigens, CD/genetics , Apoptosis/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Glycoproteins/genetics , Humans , Immunologic Surveillance/genetics , Neoplastic Stem Cells/metabolism , Peptides/genetics , Stress, Physiological
12.
Free Radic Biol Med ; 50(11): 1546-55, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21402148

ABSTRACT

Mitochondria are emerging as intriguing targets for anti-cancer agents. We tested here a novel approach, whereby the mitochondrially targeted delivery of anti-cancer drugs is enhanced by the addition of a triphenylphosphonium group (TPP(+)). A mitochondrially targeted analog of vitamin E succinate (MitoVES), modified by tagging the parental compound with TPP(+), induced considerably more robust apoptosis in cancer cells with a 1-2 log gain in anti-cancer activity compared to the unmodified counterpart, while maintaining selectivity for malignant cells. This is because MitoVES associates with mitochondria and causes fast generation of reactive oxygen species that then trigger mitochondria-dependent apoptosis, involving transcriptional modulation of the Bcl-2 family proteins. MitoVES proved superior in suppression of experimental tumors compared to the untargeted analog. We propose that mitochondrially targeted delivery of anti-cancer agents offers a new paradigm for increasing the efficacy of compounds with anti-cancer activity.


Subject(s)
Drug Delivery Systems , Mitochondria/metabolism , Organophosphorus Compounds , Proto-Oncogene Proteins c-bcl-2/metabolism , Tocopherols , Animals , Apoptosis/drug effects , Drug Therapy/trends , Humans , Jurkat Cells , Models, Animal , Molecular Targeted Therapy , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Reactive Oxygen Species/metabolism , Tocopherols/chemistry , Tocopherols/pharmacology , Transcription, Genetic/drug effects
13.
Clin Cancer Res ; 15(5): 1593-600, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19223492

ABSTRACT

PURPOSE: Vitamin E analogues are potent novel anticancer drugs. The purpose of this study was to elucidate the cellular target by which these agents, represented by alpha-tocopoheryl succinate (alpha-TOS), suppress tumors in vivo, with the focus on the mitochondrial complex II (CII). EXPERIMENTAL DESIGN: Chinese hamster lung fibroblasts with functional, dysfunctional, and reconstituted CII were transformed using H-Ras. The cells were then used to form xenografts in immunocompromized mice, and response of the cells and the tumors to alpha-TOS was studied. RESULTS: The CII-functional and CII-reconstituted cells, unlike their CII-dysfunctional counterparts, responded to alpha-TOS by reactive oxygen species generation and apoptosis execution. Tumors derived from these cell lines reciprocated their responses to alpha-TOS. Thus, growth of CII-functional and CII-reconstituted tumors was strongly suppressed by the agent, and this was accompanied by high level of apoptosis induction in the tumor cells. On the other hand, alpha-TOS did not inhibit the CII-dysfunctional tumors. CONCLUSIONS: We document in this report a novel paradigm, according to which the mitochondrial CII, which rarely mutates in human neoplasias, is a plausible target for anticancer drugs from the group of vitamin E analogues, providing support for their testing in clinical trials.


Subject(s)
Antioxidants/therapeutic use , Electron Transport Complex II/metabolism , Lung Neoplasms/prevention & control , Mitochondria/metabolism , alpha-Tocopherol/therapeutic use , Animals , Apoptosis/drug effects , Blotting, Western , Cell Transformation, Neoplastic , Cells, Cultured , Colony-Forming Units Assay , Cricetinae , Cricetulus , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Green Fluorescent Proteins/genetics , Lung/cytology , Lung/drug effects , Lung/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Proteins/physiology , Mice , Mice, Inbred BALB C , Mice, Nude , Oxygen Consumption , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
Biochem Biophys Res Commun ; 373(4): 567-71, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18590703

ABSTRACT

Recent research shows that Cancer stem cells (CSCs) are relatively resistant to apoptosis induction. We studied the effect of the immunological apoptogen TRAIL on Jurkat cells enriched in the CD133-positive population. CD133(high) Jurkat cells were more resistant to apoptosis than their CD133(low) counterparts, and showed higher level of expression of FLIP, an inhibitor of death receptor-mediated apoptosis. Breast cancer MCF7 cells showed high level of expression CD133 in the unseparated culture, with accompanying high level of FLIP. Down-regulation of FLIP by siRNA resulted in sensitisation of the cells to TRAIL, as documented by more robust apoptosis. We conclude that high expression of FLIP is at least one of the reasons for resistance of CSCs to apoptosis induced by the death ligand TRAIL.


Subject(s)
Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Glycoproteins/metabolism , Neoplastic Stem Cells/drug effects , Peptides/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , AC133 Antigen , Humans , Jurkat Cells , Neoplastic Stem Cells/metabolism , Recombinant Proteins/pharmacology , Up-Regulation
15.
Biofactors ; 34(3): 231-5, 2008.
Article in English | MEDLINE | ID: mdl-19734124

ABSTRACT

It is increasingly accepted that cancer stem cells (CSCs) are rather resistant to apoptosis to various inducers, including the immunological apoptogen TRAIL. Here we show that cancer cells with high expression of CD133, a marker that is often associated with CSCs, are resistant to TRAIL-induced apoptosis, compared to their CD133-low counterparts. We show that this resistance can be ascribed to the high expression of FLIP, an inhibitor of the extrinsic apoptotic pathway, in CD133-high cells. Downregulation of FLIP by siRNA in CD133-high cells sensitised them to TRAIL killing. Thus, CD133-high cells may be resistant to TRAIL due to high expression of FLIP.


Subject(s)
Antigens, CD/metabolism , Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Glycoproteins/metabolism , Peptides/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Up-Regulation , AC133 Antigen , Cell Line, Tumor , Flow Cytometry , Humans
16.
Cancer Res ; 67(24): 11906-13, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18089821

ABSTRACT

"Mitocans" from the vitamin E group of selective anticancer drugs, alpha-tocopheryl succinate (alpha-TOS) and its ether analogue alpha-TEA, triggered apoptosis in proliferating but not arrested endothelial cells. Angiogenic endothelial cells exposed to the vitamin E analogues, unlike their arrested counterparts, readily accumulated reactive oxygen species (ROS) by interfering with the mitochondrial redox chain and activating the intrinsic apoptotic pathway. The vitamin E analogues inhibited angiogenesis in vitro as assessed using the "wound-healing" and "tube-forming" models. Endothelial cells deficient in mitochondrial DNA (mtDNA) were resistant to the vitamin E analogues, both in ROS accumulation and apoptosis induction, maintaining their angiogenic potential. alpha-TOS inhibited angiogenesis in a mouse cancer model, as documented by ultrasound imaging. We conclude that vitamin E analogues selectively kill angiogenic endothelial cells, suppressing tumor growth, which has intriguing clinical implications.


Subject(s)
Antineoplastic Agents/therapeutic use , Mitochondria/physiology , Neovascularization, Pathologic/prevention & control , Oxidative Stress/physiology , Vitamin E/analogs & derivatives , Vitamin E/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Apoptosis/drug effects , DNA, Mitochondrial/genetics , Drug Resistance , Endothelium, Vascular , Humans , Mitochondria/drug effects , Oxidative Stress/drug effects
17.
Cancer Res ; 67(7): 3337-44, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17409443

ABSTRACT

Overexpression of erbB2 is associated with resistance to apoptosis. We explored whether high level of erbB2 expression by cancer cells allows their targeting using an erbB2-binding peptide (LTVSPWY) attached to the proapoptotic alpha-tocopheryl succinate (alpha-TOS). Treating erbB2-low or erbB2-high cells with alpha-TOS induced similar levels of apoptosis, whereas alpha-TOS-LTVSPWY induced greater levels of apoptosis in erbB2-high cells. alpha-TOS rapidly accumulated in erbB2-high cells exposed to alpha-TOS-LTVSPWY. The extent of apoptosis induced in erbB2-high cells by alpha-TOS-LTVSPWY was suppressed by erbB2 RNA interference as well as by inhibition of either endocytotic or lysosomal function. alpha-TOS-LTVSPWY reduced erbB2-high breast carcinomas in FVB/N c-neu transgenic mice. We conclude that a conjugate of a peptide targeting alpha-TOS to erbB2-overexpressing cancer cells induces rapid apoptosis and efficiently suppresses erbB2-positive breast tumors.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Oligopeptides/pharmacokinetics , Receptor, ErbB-2/biosynthesis , Vitamin E/analogs & derivatives , Breast Neoplasms/enzymology , Cell Line, Tumor , Humans , Oligopeptides/administration & dosage , Protein Binding , Receptor, ErbB-2/metabolism , Tocopherols , Vitamin E/administration & dosage , Vitamin E/pharmacokinetics
18.
Biochem Biophys Res Commun ; 355(4): 855-9, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17307142

ABSTRACT

Recent evidence suggests that a subset of cells within a tumour have 'stem-like' characteristics. These tumour-initiating cells, distinct from non-malignant stem cells, show low proliferative rates, high self-renewing capacity, propensity to differentiate into actively proliferating tumour cells, resistance to chemotherapy or radiation, and they are often characterised by elevated expression of the stem cell surface marker CD133. Understanding the molecular biology of the CD133(+) cancer cells is now essential for developing more effective cancer treatments. These may include drugs targeting organelles, such as mitochondria or lysosomes, using highly efficient and selective inducers of apoptosis. Alternatively, agents or treatment regimens that enhance sensitivity of these therapy-resistant "tumour stem cells" to the current or emerging anti-tumour drugs would be of interest as well.


Subject(s)
Antigens, CD/metabolism , Glycoproteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Peptides/metabolism , AC133 Antigen , Animals , Antigens, CD/genetics , Biomarkers, Tumor , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Glycoproteins/genetics , Humans , Neoplasms/genetics , Neoplastic Stem Cells/pathology , Peptides/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...