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










Publication year range
2.
Oncotarget ; 6(4): 2315-30, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25537513

ABSTRACT

Basal-like breast cancer is an aggressive tumor subtype with a poor response to conventional therapies. Tumor formation and relapse are sustained by a cell subset of Breast Cancer Stem Cells (BrCSCs). Here we show that miR-100 inhibits maintenance and expansion of BrCSCs in basal-like cancer through Polo-like kinase1 (Plk1) down-regulation. Moreover, miR-100 favors BrCSC differentiation, converting a basal like phenotype into luminal. It induces the expression of a functional estrogen receptor (ER) and renders basal-like BrCSCs responsive to hormonal therapy. The key role played by miR-100 in breast cancer free-survival is confirmed by the analysis of a cohort of patients' tumors, which shows that low expression of miR-100 is a negative prognostic factor and is associated with gene signatures of high grade undifferentiated tumors. Our findings indicate a new possible therapeutic strategy, which could make aggressive breast cancers responsive to standard treatments.


Subject(s)
Breast Neoplasms/genetics , Cell Differentiation/genetics , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Kaplan-Meier Estimate , MCF-7 Cells , Mice, Inbred NOD , Mice, SCID , Middle Aged , Prognosis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tamoxifen/pharmacology , Transplantation, Heterologous , Polo-Like Kinase 1
3.
Cell Stem Cell ; 14(3): 342-56, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24607406

ABSTRACT

Cancer stem cells drive tumor formation and metastasis, but how they acquire metastatic traits is not well understood. Here, we show that all colorectal cancer stem cells (CR-CSCs) express CD44v6, which is required for their migration and generation of metastatic tumors. CD44v6 expression is low in primary tumors but demarcated clonogenic CR-CSC populations. Cytokines hepatocyte growth factor (HGF), osteopontin (OPN), and stromal-derived factor 1α (SDF-1), secreted from tumor associated cells, increase CD44v6 expression in CR-CSCs by activating the Wnt/ß-catenin pathway, which promotes migration and metastasis. CD44v6(-) progenitor cells do not give rise to metastatic lesions but, when treated with cytokines, acquire CD44v6 expression and metastatic capacity. Importantly, phosphatidylinositol 3-kinase (PI3K) inhibition selectively killed CD44v6 CR-CSCs and reduced metastatic growth. In patient cohorts, low levels of CD44v6 predict increased probability of survival. Thus, the metastatic process in colorectal cancer is initiated by CSCs through the expression of CD44v6, which is both a functional biomarker and therapeutic target.


Subject(s)
Biomarkers, Tumor/metabolism , Cellular Reprogramming , Colonic Neoplasms/pathology , Hyaluronan Receptors/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Animals , Bone Morphogenetic Proteins/metabolism , Carcinogenesis/pathology , Colonic Neoplasms/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Mice, SCID , Neoplasm Metastasis , Neoplasm Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Prognosis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , Treatment Outcome , Wnt Proteins/metabolism
4.
Cancer Res ; 73(21): 6393-400, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24008319

ABSTRACT

Recombinant erythropoietin (EPO) analogs [erythropoiesis-stimulating agents (ESA)] are clinically used to treat anemia in patients with cancer receiving chemotherapy. After clinical trials reporting increased adverse events and/or reduced survival in ESA-treated patients, concerns have been raised about the potential role of ESAs in promoting tumor progression, possibly through tumor cell stimulation. However, evidence is lacking on the ability of EPO to directly affect cancer stem-like cells, which are thought to be responsible for tumor progression and relapse. We found that breast cancer stem-like cells (BCSC) isolated from patient tumors express the EPO receptor and respond to EPO treatment with increased proliferation and self-renewal. Importantly, EPO stimulation increased BCSC resistance to chemotherapeutic agents and activated cellular pathways responsible for survival and drug resistance. Specifically, the Akt and ERK pathways were activated in BCSC at early time points following EPO treatment, whereas Bcl-xL levels increased at later times. In vivo, EPO administration counteracted the effects of chemotherapeutic agents on BCSC-derived orthotopic tumor xenografts and promoted metastatic progression both in the presence and in the absence of chemotherapy treatment. Altogether, these results indicate that EPO acts directly on BCSC by activating specific survival pathways, resulting in BCSC protection from chemotherapy and enhanced tumor progression.


Subject(s)
Anemia/drug therapy , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , Erythropoietin/pharmacology , Neoplastic Stem Cells/drug effects , Anemia/chemically induced , Anemia/pathology , Animals , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Progression , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction/drug effects , Tumor Cells, Cultured
5.
Expert Opin Ther Targets ; 16(3): 259-67, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22385077

ABSTRACT

INTRODUCTION: Recent evidence based on cancer stem cell (CSC) models, is boosting the progress of translational research and providing relevant clinical implications in many tumour types, including colorectal cancer. The current failure of standard therapies is attributed to a small fraction of the primary cell population with stem-like characteristics, such as self-renewal and differentiation. Identification of CSCs is based on two different criteria of selection: stemness-selective conditions and direct isolation based on putative stem cell markers expression. CD133, a transmembrane glycoprotein, was associated with tumor-initiating cells derived from several histological variants of tumors, including colon. AREAS COVERED: In this review the current understandings about CD133 as putative marker of tumour-initiating cells in colorectal cancer (CRC) is described. The focus of the discussion is on the need for additional markers to better identify the cell population able to recapitulate the parental tumor in immunocompromised mice. EXPERT OPINION: Identification and characterization of CSCs represents a relevant issue to define innovative therapeutic approaches, overcoming the emergence of cancer cell clones capable of evading standard therapy.


Subject(s)
Antigens, CD/metabolism , Colorectal Neoplasms/metabolism , Glycoproteins/metabolism , Peptides/metabolism , AC133 Antigen , Animals , Humans , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism
6.
Gastroenterology ; 140(1): 297-309, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20951698

ABSTRACT

BACKGROUND & AIMS: The limited clinical response observed in many patients with colorectal cancer may be related to the presence of chemoresistant colorectal cancer stem cells (CRC-SCs). Bone morphogenetic protein 4 (BMP4) promotes the differentiation of normal colonic stem cells. We investigated whether BMP4 might be used to induce differentiation of CRC-SCs and for therapeutic purposes. METHODS: CRC-SCs were isolated from 25 tumor samples based on expression of CD133 or using a selection culture medium. BMP4 expression and activity on CRC-SCs were evaluated in vitro; progeny of the stem cells were evaluated by immunofluorescence, immunoblot, and flow cytometry analyses. The potential therapeutic effect of BMP4 was assessed in immunocompromised mice after injection of CRC-SCs that responded to chemotherapy (n = 4) or that did not (n = 2). RESULTS: CRC-SCs did not express BMP4 whereas differentiated cells did. Recombinant BMP4 promoted differentiation and apoptosis of CRC-SCs in 12 of 15 independent experiments; this effect did not depend on Small Mothers against decapentaplegic (Smad)4 expression level or microsatellite stability. BMP4 activated the canonical and noncanonical BMP signaling pathways, including phosphoInositide 3-kinase (PI3K) and PKB (protein kinase B)/AKT. Mutations in PI3K or loss of Phosphatase and Tensin homolog (PTEN) in Smad4-defective tumors made CRC-SCs unresponsive to BMP4. Administration of BMP4 to immunocompromised mice with tumors that arose from CRC-SCs increased the antitumor effects of 5-fluorouracil and oxaliplatin. CONCLUSIONS: BMP4 promotes terminal differentiation, apoptosis, and chemosensitization of CRC-SCs in tumors that do not have simultaneous mutations in Smad4 and constitutive activation of PI3K. BMP4 might be developed as a therapeutic agent against cancer stem cells in advanced colorectal tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Morphogenetic Protein 4/therapeutic use , Colorectal Neoplasms/drug therapy , Fluorouracil/therapeutic use , Neoplastic Stem Cells/drug effects , Organoplatinum Compounds/therapeutic use , AC133 Antigen , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Aged , Aged, 80 and over , Animals , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation , Cells, Cultured , Colorectal Neoplasms/pathology , Female , Glycoproteins/metabolism , Humans , Male , Mice , Microsatellite Instability , Middle Aged , Mutation , Oxaliplatin , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Peptides/metabolism , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Smad4 Protein/metabolism
7.
Immunotherapy ; 3(1): 97-106, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21174560

ABSTRACT

In the last 10 years, cancer stem cells have interested the scientific community because this small tumorigenic population is also associated with tumor progression in human patients and specific targeting of cancer stem cells could be a strategy to eradicate cancers currently resistant to conventional therapy. Clinical studies have recently demonstrated that adding immune therapy to chemotherapy has survival benefits in comparison with chemotherapy alone that can sensitize tumors to immune cell-mediated killing (e.g., increasing sensitivity of tumor cells to subsequent cytotoxicity by T cells via upregulation of death receptors DR5 and Fas). However, loss of MHC molecules is often observed in cancer cells, rendering tumor cells resistant to CD8 T-cell-mediated cytotoxicity. For this reason, we review the role of other T-cell subsets, such as γδ T and NK cells that are able to efficiently recognize and kill tumor cells and that could be used in passive or active immunotherapy in cancer stem cell eradication.


Subject(s)
Colonic Neoplasms/therapy , Immunotherapy/methods , Killer Cells, Natural/immunology , Neoplastic Stem Cells/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/immunology , Humans , T-Lymphocytes/immunology
8.
Cancer Lett ; 300(2): 205-14, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21041018

ABSTRACT

Mutation of the Bcr-Abl oncoprotein is one of most frequent mechanisms by which chronic myelogenous leukemia (CML) cells become resistant to imatinib. Here, we show that treatment of cell lines harbouring wild type or mutant BCR-ABL with carboxyamidotriazole (CAI), a calcium influx and signal transduction inhibitor, inhibits cell growth, the expression of Bcr-Abl and its downstream signalling, and induces apoptosis. Moreover, we show that CAI acts by increasing intracellular ROS. Clinically significant, CAI has also inhibitory effects on T315I Bcr-Abl mutant, a mutation that causes CML cells to become insensitive to imatinib and second generation abl kinase inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Oxidation-Reduction/drug effects , Triazoles/pharmacology , Animals , Apoptosis/drug effects , Benzamides , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Imatinib Mesylate , Mice , Mutation , Piperazines/pharmacology , Pyrimidines/pharmacology , Reactive Oxygen Species/metabolism
9.
Cancers (Basel) ; 3(2): 1929-46, 2011 Apr 11.
Article in English | MEDLINE | ID: mdl-24212789

ABSTRACT

Nowadays it is reported that, similarly to other solid tumors, colorectal cancer is sustained by a rare subset of cancer stem-like cells (CSCs), which survive conventional anticancer treatments, thanks to efficient mechanisms allowing escape from apoptosis, triggering tumor recurrence. To improve patient outcomes, conventional anticancer therapies have to be replaced with specific approaches targeting CSCs. In this review we provide strong support that BMP4 is an innovative therapeutic approach to prevent colon cancer growth increasing differentiation markers expression and apoptosis. Recent data suggest that in colorectal CSCs, protection from apoptosis is achieved by interleukin-4 (IL-4) autocrine production through upregulation of antiapoptotic mediators, including survivin. Consequently, IL-4 neutralization could deregulate survivin expression and localization inducing chemosensitivity of the colon CSCs pool.

10.
Cancers (Basel) ; 3(2): 1957-74, 2011 Apr 13.
Article in English | MEDLINE | ID: mdl-24212791

ABSTRACT

It is widely accepted by the scientific community that cancer, including colon cancer, is a "stem cell disease". Until a few years ago, common opinion was that all neoplastic cells within a tumor contained tumorigenic growth capacity, but recent evidences hint to the possibility that such a feature is confined to a small subset of cancer-initiating cells, also called cancer stem cells (CSCs). Thus, malignant tumors are organized in a hierarchical fashion in which CSCs give rise to more differentiated tumor cells. CSCs possess high levels of ATP-binding cassette (ABC) transporters and anti-apoptotic molecules, active DNA-repair, slow replication capacities and they produce growth factors that confer refractoriness to antineoplastic treatments. The inefficacy of conventional therapies towards the stem cell population might explain cancer chemoresistance and the high frequency of relapse shown by the majority of tumors. Nowadays, in fact all the therapies available are not sufficient to cure patients with advanced forms of colon cancer since they target differentiated cancer cells which constitute most of the tumor mass and spare CSCs. Since CSCs are the entities responsible for the development of the tumor and represent the only cell population able to sustain tumor growth and progression, these cells represent the elective target for innovative therapies.

11.
Cancer Res ; 70(21): 8874-85, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20959469

ABSTRACT

Thyroid carcinoma is the most common endocrine malignancy and the first cause of death among endocrine cancers. We show that the tumorigenic capacity in thyroid cancer is confined in a small subpopulation of stem-like cells with high aldehyde dehydrogenase (ALDH(high)) activity and unlimited replication potential. ALDH(high) cells can be expanded indefinitely in vitro as tumor spheres, which retain the tumorigenic potential upon delivery in immunocompromised mice. Orthotopic injection of minute numbers of thyroid cancer stem cells recapitulates the behavior of the parental tumor, including the aggressive metastatic features of undifferentiated thyroid carcinomas, which are sustained by constitutive activation of cMet and Akt in thyroid cancer stem cells. The identification of tumorigenic and metastagenic thyroid cancer cells may provide unprecedented preclinical tools for development and preclinical validation of novel targeted therapies.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Cell Transformation, Neoplastic/pathology , Lung Neoplasms/secondary , Neoplastic Stem Cells/pathology , Thyroid Gland/pathology , Thyroid Neoplasms/pathology , Adenocarcinoma, Follicular/enzymology , Adenocarcinoma, Follicular/pathology , Adult , Aged , Aldehyde Dehydrogenase/genetics , Animals , Blotting, Western , Carcinoma/enzymology , Carcinoma/pathology , Carcinoma, Papillary/enzymology , Carcinoma, Papillary/pathology , Case-Control Studies , Cell Adhesion , Cell Movement , Cell Proliferation , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Lung Neoplasms/enzymology , Male , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Middle Aged , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Gland/enzymology , Thyroid Neoplasms/enzymology , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays , Young Adult
12.
Clin Cancer Res ; 16(1): 45-55, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20028757

ABSTRACT

PURPOSE: To examine the role of cancer stem cells (CSC) in mediating metastasis in inflammatory breast cancer (IBC) and the association of these cells with patient outcome in this aggressive type of breast cancer. EXPERIMENTAL DESIGN: CSCs were isolated from SUM149 and MARY-X, an IBC cell line and primary xenograft, by virtue of increased aldehyde dehydrogenase (ALDH) activity as assessed by the ALDEFLUOR assay. Invasion and metastasis of CSC populations were assessed by in vitro and mouse xenograft assays. Expression of ALDH1 was determined on a retrospective series of 109 IBC patients and this was correlated with histoclinical data. All statistical tests were two sided. Log-rank tests using Kaplan-Meier analysis were used to determine the correlation of ALDH1 expression with development of metastasis and patient outcome. RESULTS: Both in vitro and xenograft assays showed that invasion and metastasis in IBC are mediated by a cellular component that displays ALDH activity. Furthermore, expression of ALDH1 in IBC was an independent predictive factor for early metastasis and decreased survival in this patient population. CONCLUSIONS: These results suggest that the metastatic, aggressive behavior of IBC may be mediated by a CSC component that displays ALDH enzymatic activity. ALDH1 expression represents the first independent prognostic marker to predict metastasis and poor patient outcome in IBC. The results illustrate how stem cell research can translate into clinical practice in the IBC field.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Breast Neoplasms/enzymology , Inflammation/enzymology , Isoenzymes/metabolism , Neoplasm Metastasis , Neoplastic Stem Cells/enzymology , Aldehyde Dehydrogenase 1 Family , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Retinal Dehydrogenase , Treatment Outcome
13.
BMC Cell Biol ; 10: 79, 2009 Nov 02.
Article in English | MEDLINE | ID: mdl-19883508

ABSTRACT

BACKGROUND: Changes in chromosome number or structure as well as supernumerary centrosomes and multipolar mitoses are commonly observed in human tumors. Thus, centrosome amplification and mitotic checkpoint dysfunctions are believed possible causes of chromosomal instability. The Retinoblastoma tumor suppressor (RB) participates in the regulation of synchrony between DNA synthesis and centrosome duplication and it is involved in transcription regulation of some mitotic genes. Primary human fibroblasts were transfected transiently with short interfering RNA (siRNA) specific for human pRb to investigate the effects of pRb acute loss on chromosomal stability. RESULTS: Acutely pRb-depleted fibroblasts showed altered expression of genes necessary for cell cycle progression, centrosome homeostasis, kinetochore and mitotic checkpoint proteins. Despite altered expression of genes involved in the Spindle Assembly Checkpoint (SAC) the checkpoint seemed to function properly in pRb-depleted fibroblasts. In particular AURORA-A and PLK1 overexpression suggested that these two genes might have a role in the observed genomic instability. However, when they were post-transcriptionally silenced in pRb-depleted fibroblasts we did not observe reduction in the number of aneuploid cells. This finding suggests that overexpression of these two genes did not contribute to genomic instability triggered by RB acute loss although it affected cell proliferation. Acutely pRb-depleted human fibroblasts showed the presence of micronuclei containing whole chromosomes besides the presence of supernumerary centrosomes and aneuploidy. CONCLUSION: Here we show for the first time that RB acute loss triggers centrosome amplification and aneuploidy in human primary fibroblasts. Altogether, our results suggest that pRb-depleted primary human fibroblasts possess an intact spindle checkpoint and that micronuclei, likely caused by mis-attached kinetochores that in turn trigger chromosome segregation errors, are responsible for aneuploidy in primary human fibroblasts where pRb is acutely depleted.


Subject(s)
Aneuploidy , Cell Nucleus/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Retinoblastoma Protein/metabolism , Aurora Kinases , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Nucleus/genetics , Cells, Cultured , Centrosome/metabolism , Chromosomal Instability , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mitosis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Retinoblastoma Protein/genetics , Polo-Like Kinase 1
14.
J Immunol ; 182(11): 7287-96, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19454726

ABSTRACT

Colon cancer comprises a small population of cancer stem cells (CSC) that is responsible for tumor maintenance and resistant to cancer therapies, possibly allowing for tumor recapitulation once treatment stops. We previously demonstrated that such chemoresistance is mediated by autocrine production of IL-4 through the up-regulation of antiapoptotic proteins. Several innate and adaptive immune effector cells allow for the recognition and destruction of cancer precursors before they constitute the tumor mass. However, cellular immune-based therapies have not been experimented yet in the population of CSCs. Here, we show that the bisphosphonate zoledronate sensitizes colon CSCs to Vgamma9Vdelta2 T cell cytotoxicity. Proliferation and production of cytokines (TNF-alpha and IFN-gamma) and cytotoxic and apoptotic molecules (TRAIL and granzymes) were also induced after exposure of Vgamma9Vdelta2 T cells to sensitized targets. Vgamma9Vdelta2 T cell cytotoxicity was mediated by the granule exocytosis pathway and was highly dependent on isoprenoid production by of tumor cells. Moreover, CSCs recognition and killing was mainly TCR mediated, whereas NKG2D played a role only when tumor targets expressed several NKG2D ligands. We conclude that intentional activation of Vgamma9Vdelta2 T cells by zoledronate may substantially increase antitumor activities and represent a novel strategy for colon cancer immunotherapy.


Subject(s)
Colonic Neoplasms/immunology , Cytotoxicity, Immunologic/immunology , Neoplastic Stem Cells/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Chromaffin Granules , Cytokines , Diphosphonates/pharmacology , Humans , Imidazoles/pharmacology , Immunotherapy , NK Cell Lectin-Like Receptor Subfamily K/immunology , Terpenes , Zoledronic Acid
15.
Cancer Res ; 69(4): 1302-13, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19190339

ABSTRACT

Tumors may be initiated and maintained by a cellular subcomponent that displays stem cell properties. We have used the expression of aldehyde dehydrogenase as assessed by the ALDEFLUOR assay to isolate and characterize cancer stem cell (CSC) populations in 33 cell lines derived from normal and malignant mammary tissue. Twenty-three of the 33 cell lines contained an ALDEFLUOR-positive population that displayed stem cell properties in vitro and in NOD/SCID xenografts. Gene expression profiling identified a 413-gene CSC profile that included genes known to play a role in stem cell function, as well as genes such as CXCR1/IL-8RA not previously known to play such a role. Recombinant interleukin-8 (IL-8) increased mammosphere formation and the ALDEFLUOR-positive population in breast cancer cell lines. Finally, we show that ALDEFLUOR-positive cells are responsible for mediating metastasis. These studies confirm the hierarchical organization of immortalized cell lines, establish techniques that can facilitate the characterization of regulatory pathways of CSCs, and identify potential stem cell markers and therapeutic targets.


Subject(s)
Breast Neoplasms/pathology , Breast/cytology , Neoplasm Metastasis/pathology , Stem Cells/pathology , Aldehyde Dehydrogenase/genetics , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line , Cell Line, Tumor , Female , Flow Cytometry , Gene Expression Profiling , Homeostasis , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Receptors, Interleukin-8A/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Stem Cells/physiology
16.
Cancer Res ; 67(11): 5522-30, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17545635

ABSTRACT

Overexpression of MUC1 oncoprotein is frequently observed in cancer and contributes to confer resistance to genotoxic agents. Papillary, follicular, and anaplastic thyroid carcinomas are the three forms of thyroid epithelial cancer. Anaplastic tumors are less differentiated and extremely aggressive, characterized by a poor prognosis. Little is known about the role of MUC1 in thyroid cancer. We recently showed that autocrine production of interleukin (IL)-4 and IL-10 controls thyroid cancer cell survival, growth, and resistance to chemotherapy through activation of Janus-activated kinase/signal transducers and activators of transcription (JAK/STAT) and phosphatidylinositide 3'-OH kinase (PI3K)/Akt pathways. In the present study, we showed that MUC1 COOH-terminal subunit (MUC1-C) is overexpressed in all the histologic variants of thyroid cancer cells and localizes to mitochondria where it interferes with the release of mitochondrial proapoptotic proteins. Moreover, IL-4 and IL-10 promote the increase of MUC1-C expression levels in normal thyroid cells, whereas blockage of both cytokines or neutralization of JAK/STAT and PI3K/Akt pathways through the exogenous expression of SOCS-1 and Akt(K179M) leads to a significant decrease of MUC1-C in primary thyroid cancer cells. Interestingly, down-regulation of MUC1 expression by direct targeting with RNA interference sensitizes anaplastic thyroid cancer cells to chemotherapy-induced apoptosis in vitro. Thus, MUC1 is a main component of the survival network acting in thyroid cancer and could be considered a key molecular target for sensitizing cancer cells to conventional or novel treatments.


Subject(s)
Antigens, Neoplasm/biosynthesis , Mucins/biosynthesis , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/metabolism , Antibiotics, Antineoplastic/pharmacology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Apoptosis , Cell Line, Tumor , Down-Regulation , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Humans , Interleukin-10/pharmacology , Interleukin-4/pharmacology , Mitochondria/metabolism , Mucin-1 , Mucins/genetics , Mucins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/biosynthesis , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Thyroid Neoplasms/genetics , Transcription, Genetic , Transfection
17.
Cell Stem Cell ; 1(4): 389-402, 2007 Oct 11.
Article in English | MEDLINE | ID: mdl-18371377

ABSTRACT

A novel paradigm in tumor biology suggests that cancer growth is driven by stem-like cells within a tumor. Here, we describe the identification and characterization of such cells from colon carcinomas using the stem cell marker CD133 that accounts around 2% of the cells in human colon cancer. The CD133(+) cells grow in vitro as undifferentiated tumor spheroids, and they are both necessary and sufficient to initiate tumor growth in immunodeficient mice. Xenografts resemble the original human tumor maintaining the rare subpopulation of tumorigenic CD133(+) cells. Further analysis revealed that the CD133(+) cells produce and utilize IL-4 to protect themselves from apoptosis. Consistently, treatment with IL-4Ralpha antagonist or anti-IL-4 neutralizing antibody strongly enhances the antitumor efficacy of standard chemotherapeutic drugs through selective sensitization of CD133(+) cells. Our data suggest that colon tumor growth is dictated by stem-like cells that are treatment resistant due to the autocrine production of IL-4.


Subject(s)
Colonic Neoplasms/pathology , Interleukin-4/biosynthesis , Neoplastic Stem Cells/pathology , AC133 Antigen , Aged , Animals , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Female , Fluorouracil/pharmacology , Glycoproteins/metabolism , Humans , Male , Mice , Mice, Nude , Middle Aged , Neoplastic Stem Cells/drug effects , Neutralization Tests , Organoplatinum Compounds/pharmacology , Oxaliplatin , Peptides/metabolism , Receptors, Interleukin-4/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Xenograft Model Antitumor Assays
18.
Mol Cancer ; 5: 38, 2006 Sep 20.
Article in English | MEDLINE | ID: mdl-16987420

ABSTRACT

BACKGROUND: Incorrect segregation of whole chromosomes or parts of chromosome leads to aneuploidy commonly observed in cancer. The correct centrosome duplication, assuring assembly of a bipolar mitotic spindle, is essential for chromosome segregation fidelity and preventing aneuploidy. Alteration of p53 and pRb functions by expression of HPV16-E6 and E7 oncoproteins has been associated with centrosome amplification. However, these last findings could be the result of targeting cellular proteins in addition to pRb by HPV16-E7 oncoprotein. To get a more detailed picture on the role of pRb in chromosomal instability and centrosome amplification, we analyzed the effects of the acute loss of retinoblastoma gene function in primary conditional Rb deficient mouse embryonic fibroblasts (MEFs). Moreover, since pRb is a transcriptional repressor, microarray analysis was done on pRb-competent and pRb-deficient MEFs to evaluate changes in expression of genes for centrosome homeostasis and for correct mitosis. RESULTS: Acute loss of pRb induces centrosome amplification and aneuploidy in the vast majority of cells analyzed. A time course analysis shows a decrease of cells with amplified centrosomes after 40 days from the adenoviral infection. At this time only 12% of cells still show amplified centrosomes. Interestingly, cells with pRb constitutive loss show a similar percentage of cells with amplified centrosomes. DNA-Chip analyses in MEFs wt (mock infected) and pRb depleted (Ad-Cre infected) cells reveal differential expression of genes controlling both centrosome duplication and mitotic progression. CONCLUSION: Our findings suggest a direct link between pRb status, centrosome amplification and chromosomal instability, and define specific mitotic genes as targets whose gene expression has to be altered to achieve or maintain aneuploidy.


Subject(s)
Aneuploidy , Centrosome/metabolism , Gene Amplification , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/genetics , Animals , Cells, Cultured , Fibroblasts , Gene Expression , Mice , Mitosis , Retinoblastoma Protein/metabolism , Time Factors , Transcription, Genetic/genetics
19.
Cancer Lett ; 238(1): 153-60, 2006 Jul 08.
Article in English | MEDLINE | ID: mdl-16154257

ABSTRACT

Alterations in the number and/or morphology of centrosomes are frequently observed in human tumours. However, it is still debated if a direct link between supernumerary centrosomes and tumorigenesis exists and if centrosome amplification could directly cause aneuploidy. Here, we report that hydroxyurea treatment induced centrosome amplification in both human fibroblasts expressing the HPV16 -E6-E7 oncoproteins, which act principally by targeting p53 and pRB, respectively, and in conditional pRB deficient mouse fibroblasts. Following hydroxyurea removal both normal and p53 deficient human fibroblasts arrested. On the contrary pRB deficient fibroblasts entered the cell cycle generating aneuploid cells. Also the majority of conditional Rb deficient MEFs showed supernumerary centrosomes and aneuploid cells which increased over time. Finally, our results suggest that pRB dysfunction both in human and murine fibroblasts transiently arrested in G1/S by hydroxyurea allows centrosomes amplification, in the absence of DNA synthesis, that in turn could drive aneuploidy.


Subject(s)
Aneuploidy , Centrosome/drug effects , Centrosome/physiology , Fibroblasts/drug effects , Hydroxyurea/pharmacology , Retinoblastoma Protein/deficiency , Animals , Cell Line , Cells, Cultured , Chromosomal Instability/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Mice , Retinoblastoma Protein/genetics
20.
Arthritis Rheum ; 52(7): 2179-91, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15986355

ABSTRACT

OBJECTIVE: To determine whether prolongation of the inflammatory reaction in patients with Behçet's disease (BD) is related to apoptosis resistance and is associated with the up-regulation of antiapoptotic factors. METHODS: The percentage of cell death was evaluated by flow cytometry in peripheral blood mononuclear cells from 35 patients with BD and 30 healthy volunteers. The expression levels of antiapoptotic factors and NF-kappaB regulatory proteins were measured using Western blotting and immunohistochemical analyses. To down-regulate NF-kappaB nuclear translocation, BD T lymphocytes were exposed in vitro to thalidomide and subjected to transfection with NF-kappaB small interfering RNA. RESULTS: Although CD95 is highly expressed in BD T cells, the absence of sensitivity to CD95-induced apoptosis observed may be attributable to the inhibitory action of antiapoptotic genes. Immunoblot analysis for major antiapoptotic proteins showed considerable up-regulation of the short form of cellular FLIP (cFLIP) and Bcl-x(L) in BD activated T cells, while levels of Bcl-2, caspase 3, and caspase 8 in activated T cells from patients with BD were comparable with those in activated T cells from normal donors. Moreover, expression of IKK and IkappaB was up-regulated, whereas NF-kappaB translocated to the nucleus in BD T cells, suggesting that NF-kappaB activation may modulate the expression of antiapoptotic genes. Interestingly, thalidomide and NF-kappaB small interfering RNA down-regulated cFLIP and Bcl-x(L) expression levels and sensitized BD activated T cells to CD95-induced apoptosis. CONCLUSION: Taken together, these results indicate that NF-kappaB contributes to the regulation of the apoptosis-related factors and death receptors leading to apoptosis resistance in BD T cell subsets. Our results suggest that NF-kappaB plays a crucial role in the pathogenesis of BD, and that its pharmacologic control could represent a key strategy in modulating specific immune-mediated disease.


Subject(s)
Apoptosis/immunology , Behcet Syndrome/immunology , Intracellular Signaling Peptides and Proteins/immunology , NF-kappa B/immunology , Proto-Oncogene Proteins c-bcl-2/immunology , T-Lymphocytes/immunology , fas Receptor/immunology , Adult , Behcet Syndrome/metabolism , Behcet Syndrome/pathology , CASP8 and FADD-Like Apoptosis Regulating Protein , Cells, Cultured , Dose-Response Relationship, Drug , Female , Flow Cytometry , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Male , NF-kappa B/biosynthesis , NF-kappa B/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Thalidomide/pharmacology , Transfection , Up-Regulation , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...