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










Publication year range
1.
Cancer Res ; 80(11): 2311-2324, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32179512

ABSTRACT

Tumor growth and development is determined by both cancer cell-autonomous and microenvironmental mechanisms, including the contribution of infiltrating immune cells. Because the role of mast cells (MC) in this process is poorly characterized and even controversial, we investigated their part in breast cancer. Crossing C57BL/6 MMTV-PyMT mice, which spontaneously develop mammary carcinomas, with MC-deficient C57BL/6-KitW-sh/W-sh (Wsh) mice, showed that MCs promote tumor growth and prevent the development of basal CK5-positive areas in favor of a luminal gene program. When cocultured with breast cancer cells in vitro, MCs hindered activation of cMET, a master regulator of the basal program, and simultaneously promoted expression and activation of estrogen receptor (ESR1/ER) and its target genes (PGR, KRT8/CK8, BCL2), which are all luminal markers. Moreover, MCs reduced ERBB2/HER2 levels, whose inhibition further increased ESR1 expression. In vivo and in silico analysis of patients with breast cancer revealed a direct correlation between MC density and ESR1 expression. In mice engrafted with HER2-positive breast cancer tumors, coinjection of MCs increased tumor engraftment and outgrowth, supporting the link between MCs and increased risk of relapse in patients with breast cancer. Together, our findings support the notion that MCs influence the phenotype of breast cancer cells by stimulating a luminal phenotype and ultimately modifying the outcome of the disease. SIGNIFICANCE: Mast cells impact breast cancer outcome by directly affecting the phenotype of tumor cells through stimulation of the estrogen receptor pathway.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Mast Cells/pathology , Receptors, Estrogen/metabolism , Animals , Breast Neoplasms/genetics , Cell Communication/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , ErbB Receptors/metabolism , Female , Humans , Male , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis , Proto-Oncogene Proteins c-met/metabolism , Receptor, ErbB-2/metabolism
2.
Cell Death Differ ; 25(12): 2147-2164, 2018 12.
Article in English | MEDLINE | ID: mdl-29674627

ABSTRACT

Inhibitor of apoptosis (IAP) proteins constitute a family of conserved molecules that regulate both apoptosis and receptor signaling. They are often deregulated in cancer cells and represent potential targets for therapy. In our work, we investigated the effect of IAP inhibition in vivo to identify novel downstream genes expressed in an IAP-dependent manner that could contribute to cancer aggressiveness. To this end, immunocompromised mice engrafted subcutaneously with the triple-negative breast cancer MDA-MB231 cell line were treated with SM83, a Smac mimetic that acts as a pan-IAP inhibitor, and tumor nodules were profiled for gene expression. SM83 reduced the expression of Snai2, an epithelial-to-mesenchymal transition factor often associated with increased stem-like properties and metastatic potential especially in breast cancer cells. By testing several breast cancer cell lines, we demonstrated that Snai2 downregulation prevents cell motility and that its expression is promoted by cIAP1. In fact, the chemical or genetic inhibition of cIAP1 blocked epidermal growth factor receptor (EGFR)-dependent activation of the mitogen-activated protein kinase (MAPK) pathway and caused the reduction of Snai2 transcription levels. In a number of breast cancer cell lines, cIAP1 depletion also resulted in a reduction of EGFR protein levels which derived from the decrease of its gene transcription, though, paradoxically, the silencing of cIAP1 promoted EGFR protein stability rather than its degradation. Finally, we provided evidence that IAP inhibition displays an anti-tumor and anti-metastasis effect in vivo. In conclusion, our work indicates that IAP-targeted therapy could contribute to EGFR inhibition and to the reduction of its downstream mediators. This approach could be particularly effective in tumors characterized by high levels of EGFR and Snai2, such as triple-negative breast cancer.


Subject(s)
Inhibitor of Apoptosis Proteins/metabolism , Snail Family Transcription Factors/metabolism , Triple Negative Breast Neoplasms/metabolism , Administration, Intravenous , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/deficiency , Injections, Intraperitoneal , Mice , Mice, Inbred NOD , Mice, SCID , Oligopeptides/administration & dosage , Oligopeptides/pharmacology , Snail Family Transcription Factors/antagonists & inhibitors , Snail Family Transcription Factors/genetics , Triple Negative Breast Neoplasms/drug therapy , Tumor Cells, Cultured
3.
J Hematol Oncol ; 11(1): 45, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29558956

ABSTRACT

BACKGROUND: Fibroblasts are crucial mediators of tumor-stroma cross-talk through synthesis and remodeling of the extracellular matrix and production of multiple soluble factors. Nonetheless, little is still known about specific determinants of fibroblast pro-tumorigenic activity in lung cancer. Here, we aimed at understanding the role of miRNAs, which are often altered in stromal cells, in reprogramming fibroblasts towards a tumor-supporting phenotype. METHODS: We employed a co-culture-based high-throughput screening to identify specific miRNAs modulating the pro-tumorigenic potential of lung fibroblasts. Multiplex assays and ELISA were instrumental to study the effect of miRNAs on the secretome of both primary and immortalized lung fibroblasts from lung cancer patients and to evaluate plasmatic levels of HGF in heavy smokers. Direct mRNA targeting by miRNAs was investigated through dual-luciferase reporter assay and western blot. Finally, the pro-tumorigenic activity of fibroblasts and their conditioned media was tested by employing in vitro migration experiments and mouse xenografts. RESULTS: We identified miR-16 as a master regulator of fibroblast secretome and showed that its upregulation reduces HGF secretion by fibroblasts, impairing their capacity to promote cancer cell migration. This effect is due to a pleiotropic activity of miR-16 which prevents HGF expression through direct inhibition of FGFR-1 signaling and targeting of HGF mRNA. Mechanistically, miR-16 targets FGFR-1 downstream mediator MEK1, thus reducing ERK1/2 activation. Consistently, chemical or genetic inhibition of FGFR-1 mimics miR-16 activity and prevents HGF production. Of note, we report that primary fibroblast cell lines derived from lungs of heavy smokers express reduced miR-16 levels compared to those from lungs not exposed to smoke and that HGF concentration in heavy smokers' plasma correlates with levels of tobacco exposure. Finally, in vivo experiments confirmed that restoration of miR-16 expression in fibroblasts reduced their ability to promote tumor growth and that HGF plays a central role in the pro-tumorigenic activity of fibroblasts. CONCLUSIONS: Overall, these results uncover a central role for miR-16 in regulating HGF production by lung fibroblasts, thus affecting their pro-tumorigenic potential. Correlation between smoking exposure and miR-16 levels could provide novel clues regarding the formation of a tumor-proficient milieu during the early phases of lung cancer development.


Subject(s)
Fibroblasts/metabolism , Hepatocyte Growth Factor/antagonists & inhibitors , Lung/metabolism , MicroRNAs/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Female , Lung/pathology , Mice
4.
Cancer Lett ; 389: 59-69, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28040547

ABSTRACT

Using a high-throughput approach, we identified lemur tyrosine kinase 2 (LMTK2) as a novel determinant of cell sensitivity to TRAIL. LMTK2 is a poorly characterized serine/threonine kinase believed to play a role in endosomal membrane trafficking and neuronal physiology, and recently found to be mutated in diverse tumor types. We show that LMTK2 silencing sensitizes immortalized epithelial cells and cancer cells to TRAIL, and this phenomenon is accompanied by changes in the expression of BCL2 family members. In epithelial cells, LMTK2 targeting causes the down-regulation of the BCL2 and BCL-xL anti-apoptotic proteins and the reciprocal up-regulation of the pro-apoptotic protein BIM, while, in cancer cells, LMTK2 knock-down reduces BCL2 without increasing BIM levels. We provide evidence that both BIM and BCL2 proteins are regulated by LMTK2 in a GSK3ß- and PP1A-dependent manner and that their perturbation, together with BCL-xL reduction, determines an increased sensitivity not only to TRAIL, but also to other compounds. Overall, our findings suggest a broad function of LMTK2 in the regulation of the apoptotic pathway and highlight LMTK2 as a novel candidate target to increase the cytotoxic activity of chemotherapeutic compounds.


Subject(s)
Apoptosis/drug effects , Membrane Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-bcl-2/analysis , bcl-X Protein/analysis , Bcl-2-Like Protein 11/analysis , Cell Line, Tumor , ErbB Receptors/analysis , Extracellular Signal-Regulated MAP Kinases/physiology , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Membrane Proteins/antagonists & inhibitors , Protein Phosphatase 1/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA, Small Interfering/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology
5.
Oncotarget ; 6(19): 17817-31, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-26158765

ABSTRACT

Cell cycle and apoptosis regulator 2 (CCAR2, formerly known as DBC1) is a nuclear protein largely involved in DNA damage response, apoptosis, metabolism, chromatin structure and transcription regulation. Upon DNA lesions, CCAR2 is phosphorylated by the apical kinases ATM/ATR and this phosphorylation enhances CCAR2 binding to SIRT1, leading to SIRT1 inhibition, p53 acetylation and p53-dependent apoptosis. Recently, we found that also the checkpoint kinase Chk2 and the proteasome activator REGγ are required for efficient CCAR2-mediated inhibition of SIRT1 and induction of p53-dependent apoptosis.Here, we report that CCAR2 is required for the repair of heterochromatic DNA lesions, as cells knock-out for CCAR2 retain, at late time-points after genotoxic treatment, abnormal levels of DNA damage-associated nuclear foci, whose timely resolution is reinstated by HP1ß depletion. Conversely, repair of DNA damages in euchromatin are not affected by CCAR2 absence.We also report that the impairment in heterochromatic DNA repair is caused by defective Chk2 activation, detectable in CCAR2 ablated cells, which finally impacts on the phosphorylation of the Chk2 substrate KAP1 that is required for the induction of heterochromatin relaxation and DNA repair.These studies further extend and confirm the role of CCAR2 in the DNA damage response and DNA repair and illustrate a new mechanism of Chk2 activity regulation. Moreover, the involvement of CCAR2 in the repair of heterochromatic DNA breaks suggests a new role for this protein in the maintenance of chromosomal stability, which is necessary to prevent cancer formation.


Subject(s)
Checkpoint Kinase 2/metabolism , DNA Damage/physiology , DNA Repair/physiology , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Blotting, Western , Cell Cycle Proteins , Cell Line, Tumor , Chromobox Protein Homolog 5 , Fluorescent Antibody Technique , Gene Knockout Techniques , Humans , Immunoprecipitation , Nerve Tissue Proteins , Phosphorylation , RNA, Small Interfering , Transfection , Tripartite Motif-Containing Protein 28
6.
Nucleic Acids Res ; 42(21): 13150-60, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25361978

ABSTRACT

Human DBC1 (Deleted in Breast Cancer 1; KIAA1967; CCAR2) is a protein implicated in the regulation of apoptosis, transcription and histone modifications. Upon DNA damage, DBC1 is phosphorylated by ATM/ATR on Thr454 and this modification increases its inhibitory interaction with SIRT1, leading to p53 acetylation and p53-dependent apoptosis. Here, we report that the inhibition of SIRT1 by DBC1 in the DNA damage response (DDR) also depends on Chk2, the transducer kinase that is activated by ATM upon DNA lesions and contributes to the spreading of DNA damage signal. Indeed we found that inactivation of Chk2 reduces DBC1-SIRT1 binding, thus preventing p53 acetylation and DBC1-induced apoptosis. These events are mediated by Chk2 phosphorylation of the 11S proteasome activator REGγ on Ser247, which increases REGγ-DBC1 interaction and SIRT1 inhibition. Overall our results clarify the mechanisms underlying the DBC1-dependent SIRT1 inhibition and link, for the first time, Chk2 and REGγ to the ATM-DBC1-SIRT1 axis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Autoantigens/metabolism , Checkpoint Kinase 2/metabolism , DNA Damage , Proteasome Endopeptidase Complex/metabolism , Sirtuin 1/metabolism , Cell Line, Tumor , Humans , Sirtuin 1/antagonists & inhibitors
7.
J Mol Cell Biol ; 4(5): 294-303, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22735644

ABSTRACT

Human DBC1 (deleted in breast cancer-1; KIAA1967) is a nuclear protein that, in response to DNA damage, competitively inhibits the NAD(+)-dependent deacetylase SIRT1, a regulator of p53 apoptotic functions in response to genotoxic stress. DBC1 depletion in human cells increases SIRT1 activity, resulting in the deacetylation of p53 and protection from apoptosis. However, the mechanisms regulating this process have not yet been determined. Here, we report that, in human cell lines, DNA damage triggered the phosphorylation of DBC1 on Thr454 by ATM (ataxia telangiectasia-mutated) and ATR (ataxia telangiectasia and Rad3-related) kinases. Phosphorylated DBC1 bound to and inhibited SIRT1, resulting in the dissociation of the SIRT1-p53 complex and stimulating p53 acetylation and p53-dependent cell death. Indeed, DBC1-mediated genotoxicity, which was shown in knockdown experiments to be dependent on SIRT1 and p53 expression, was defective in cells expressing the phospho-mutant DBC1(T454A). This study describes the first post-translational modification of DBC1 and provides new mechanistic insight linking ATM/ATR to the DBC1-SIRT1-p53 apoptotic axis triggered by DNA damage.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Sirtuin 1/antagonists & inhibitors , Acetylation , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Line, Tumor , DNA Damage/physiology , HEK293 Cells , Humans , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Sirtuin 1/genetics , Sirtuin 1/metabolism
8.
Biochim Biophys Acta ; 1803(10): 1213-23, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20599567

ABSTRACT

The checkpoint kinase Chk2 is an effector component of the ATM-dependent DNA damage response (DDR) pathway. The activation of Chk2 by genotoxic stress involves its phosphorylation on T68 by ATM and additional auto/transphosphorylations. Here we demonstrate that in unperturbed cells, chemical inhibition of Chk2 by VRX0466617 (VRX) enhances the phosphorylation of Chk2-T68 throughout the cell cycle phases. This event, dependent on the presence of ATM and catalytically functional Chk2, is not consequential to DNA damage, as neither gamma-H2AX nuclear foci nor increased ATM activation is detected in VRX-treated cells, suggesting the involvement of other regulatory proteins. As serine/threonine protein phosphatases (PPs) regulate the phosphorylation and deactivation of proteins of the DDR pathway, we analyzed their role in phospho-T68-Chk2 regulation. We found that intracellular inhibition of PP1 and PP2A-like activities by okadaic acid markedly raised the accumulation of Chk2-pT68 without DNA damage induction, and this phenomenon was also seen when PP1-C, PP2A-C, and Wip1/PPM1D were simultaneously knockdown by siRNA. Altogether, these data indicate a novel mechanism in undamaged cells where PPs function to maintain the balance between ATM and its direct substrate Chk2 through a regulatory circuit.


Subject(s)
DNA Damage , Feedback, Physiological , Phosphoprotein Phosphatases/metabolism , Protein Serine-Threonine Kinases/metabolism , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Tumor , Checkpoint Kinase 2 , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Humans , Models, Biological , Okadaic Acid/pharmacology , Phosphoprotein Phosphatases/genetics , Phosphorylation/drug effects , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Phosphatase 2C , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA Interference , Thiazoles/pharmacology , Threonine/metabolism , Tumor Suppressor Proteins/metabolism
9.
Proc Natl Acad Sci U S A ; 106(38): 16281-6, 2009 Sep 22.
Article in English | MEDLINE | ID: mdl-19805294

ABSTRACT

The identification of lung tumor-initiating cells and associated markers may be useful for optimization of therapeutic approaches and for predictive and prognostic information in lung cancer patients. CD133, a surface glycoprotein linked to organ-specific stem cells, was described as a marker of cancer-initiating cells in different tumor types. Here, we report that a CD133+, epithelial-specific antigen-positive (CD133+ESA+) population is increased in primary nonsmall cell lung cancer (NSCLC) compared with normal lung tissue and has higher tumorigenic potential in SCID mice and expression of genes involved in stemness, adhesion, motility, and drug efflux than the CD133(-) counterpart. Cisplatin treatment of lung cancer cells in vitro resulted in enrichment of CD133+ fraction both after acute cytotoxic exposure and in cells with stable cisplatin-resistant phenotype. Subpopulations of CD133+ABCG2+ and CD133+CXCR4+ cells were spared by in vivo cisplatin treatment of lung tumor xenografts established from primary tumors. A tendency toward shorter progression-free survival was observed in CD133+ NSCLC patients treated with platinum-containing regimens. Our results indicate that chemoresistant populations with highly tumorigenic and stem-like features are present in lung tumors. The molecular features of these cells may provide the rationale for more specific therapeutic targeting and the definition of predictive factors in clinical management of this lethal disease.


Subject(s)
Antigens, CD/metabolism , Cisplatin/pharmacology , Glycoproteins/metabolism , Lung Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Peptides/metabolism , AC133 Antigen , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/therapeutic use , Drug Resistance, Neoplasm , Female , Flow Cytometry , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, SCID , Middle Aged , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptors, CXCR4/metabolism , Survival Analysis , Tumor Burden/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Cell Cycle ; 8(15): 2399-407, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19556897

ABSTRACT

REGgamma (also called PA28gamma or PSME3) is a proteasome activator involved in the degradation of several proteins that regulate cell cycle and transcription. Recently, we demonstrated that this protein has a role also in the maintenance of chromosomal stability and in the response to spindle damaging agents. Here we report for the first time that REGgamma interacts with the promyelocytic leukemia protein (PML), accumulates in PML nuclear bodies (PML-NBs), but it does not play any role in normal or arsenic-induced PML degradation. However, REGgamma seems to regulate PML-NBs number, since its deficiency causes an increase in PML-NBs, which can be overcome by increased levels of SUMO1, and its overexpression has the opposite effect. We additionally found that REGgamma interacts with the DNA damage checkpoint kinase Chk2, whose presence is necessary for the increase of PML-NBs induced by REGgamma deficiency, and that REGgamma depletion resulted in a partial restoration of PML-NBs in APL derived cells. Altogether, these results underline a new role for REGgamma in the control and regulation of PML subnuclear structures.


Subject(s)
Autoantigens/metabolism , Cell Nucleus/enzymology , Intranuclear Inclusion Bodies/metabolism , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/ultrastructure , Checkpoint Kinase 2 , Fibroblasts/enzymology , Humans , Intranuclear Inclusion Bodies/ultrastructure , Promyelocytic Leukemia Protein , SUMO-1 Protein/metabolism
11.
Curr Biol ; 19(10): 874-9, 2009 May 26.
Article in English | MEDLINE | ID: mdl-19375317

ABSTRACT

The shelterin complex [1] shapes and protects telomeric DNA from being processed as double strand breaks (DSBs) [2, 3]. Here we show that in human undamaged cells, a fraction of the kinase Chk2, a downstream target of ATM and mediator of checkpoint responses and senescence [4, 5], physically interacts with the shelterin subunit TRF2 and colocalizes with this complex at chromosome ends. This interaction, enhanced by TRF2 binding to telomeric DNA, inhibits the activation and senescence-induced function of Chk2 by a mechanism in which TRF2 binding to the N terminus of Chk2 surrounding Thr68 hinders the phosphorylation of this priming site. In response to radiation-induced DSBs, but not chromatin-remodelling agents, the telomeric Chk2-TRF2 binding dissociates in a Chk2 activity-dependent manner. Moreover, active Chk2 phosphorylates TRF2 and decreases its binding to telomeric DNA repeats, corroborating the evidences on the specific TRF2 relocalization in presence of DSBs [6]. Altogether, the capacity of TRF2 to locally repress Chk2 provides an additional level of control by which shelterin restrains the DNA damage response from an unwanted activation [6, 7] and may explain why TRF2 overexpression acts as a telomerase-independent oncogenic stimulus [8].


Subject(s)
DNA Damage , Protein Serine-Threonine Kinases/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 2/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cellular Senescence , Checkpoint Kinase 2 , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Neoplasms/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Telomerase/metabolism , Telomeric Repeat Binding Protein 2/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
12.
Clin Cancer Res ; 15(6): 2010-21, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19276287

ABSTRACT

PURPOSE: The existence of tumor-initiating cells in breast cancer has profound implications for cancer therapy. In this study, we investigated the sensitivity of tumor-initiating cells isolated from human epidermal growth factor receptor type 2 (HER2)-overexpressing carcinoma cell lines to trastuzumab, a compound used for the targeted therapy of breast cancer. EXPERIMENTAL DESIGN: Spheres were analyzed by indirect immunofluorescence for HER2 cell surface expression and by real-time PCR for HER2 mRNA expression in the presence or absence of the Notch1 signaling inhibitor (GSI) or Notch1 small interfering RNA. Xenografts of HER2-overexpressing breast tumor cells were treated with trastuzumab or doxorubicin. The sphere-forming efficiency (SFE) and serial transplantability of tumors were assessed. RESULTS: In HER2-overexpressing carcinoma cell lines, cells with tumor-initiating cell properties presented increased HER2 levels compared with the bulk cell population without modification in HER2 gene amplification. HER2 levels were controlled by Notch1 signaling, as shown by the reduction of HER2 cell surface expression and lower SFE following gamma-secretase inhibition or Notch1 specific silencing. We also show that trastuzumab was able to effectively target tumor-initiating cells of HER2-positive carcinoma cell lines, as indicated by the significant decrease in SFE and the loss of serial transplantability, following treatment of HER2-overexpressing xenotransplants. CONCLUSIONS: Here, we provide evidence for the therapeutic efficacy of trastuzumab in debulking and in targeting tumor-initiating cells of HER2-overexpressing tumors. We also propose that Notch signaling regulates HER2 expression, thereby representing a critical survival pathway of tumor-initiating cells.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/analysis , Animals , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Humans , Lapatinib , Mice , Quinazolines/pharmacology , RNA, Messenger/analysis , Receptor, ErbB-2/genetics , Receptor, Notch1/physiology , Trastuzumab , Xenograft Model Antitumor Assays
13.
Cell Cycle ; 7(4): 504-12, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18235248

ABSTRACT

REGgamma is a member of the 11S regulatory particle that activates the 20S proteasome. Studies in REGgamma deficient mice indicated an additional role for this protein in cell cycle regulation and proliferation control. In this paper we demonstrate that REGgamma protein is equally expressed throughout the cell cycle, but undergoes a distinctive subcellular localization at mitosis. Thus, while in interphase cells REGgamma is nuclear, in telophase cells it localizes on chromosomes, suggesting a role in mitotic progression. Furthermore, we found that REGgamma overexpression weakens the mitotic arrest induced by spindle damage, allowing premature exit from mitosis, whereas REGgamma depletion has the opposite effect, thus reflecting a new REGgamma function, unrelated to its role as proteasome activator. Additionally, we found that primary cells from REGgamma-/- mice and human fibroblasts with depleted expression of REGgamma or overexpressing a dominant negative mutant unable to activate the 20S proteasome, demonstrated a marked aneuploidy (chromosomal gains and losses), supernumerary centrosomes and multipolar spindles. These findings thus underscore a previously uncharacterized function of REGgamma in centrosome and chromosomal stability maintenance.


Subject(s)
Autoantigens/metabolism , Centrosome/metabolism , Chromosomal Instability/physiology , Mitosis/physiology , Proteasome Endopeptidase Complex/metabolism , Animals , Autoantigens/genetics , Autoantigens/physiology , Blotting, Western , Chromosomal Instability/genetics , DNA Primers/genetics , Fibroblasts , Flow Cytometry , Humans , Mice , Mice, Knockout , Microscopy, Fluorescence , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/physiology
14.
Mol Cell Biol ; 26(21): 7832-45, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16940182

ABSTRACT

Chk2 kinase is activated by DNA damage to regulate cell cycle arrest, DNA repair, and apoptosis. Phosphorylation of Chk2 in vivo by ataxia telangiectasia-mutated (ATM) on threonine 68 (T68) initiates a phosphorylation cascade that promotes the full activity of Chk2. We identified three serine residues (S19, S33, and S35) on Chk2 that became phosphorylated in vivo rapidly and exclusively in response to ionizing radiation (IR)-induced DNA double-strand breaks in an ATM- and Nbs1-dependent but ataxia telangiectasia- and Rad3-related-independent manner. Phosphorylation of these residues, restricted to the G(1) phase of the cell cycle, was induced by a higher dose of IR (>1 Gy) than that required for phosphorylation of T68 (0.25 Gy) and declined by 45 to 90 min, concomitant with a rise in Chk2 autophosphorylation. Compared to the wild-type form, Chk2 with alanine substitutions at S19, S33, and S35 (Chk2(S3A)) showed impaired dimerization, defective auto- and trans-phosphorylation activities, and reduced ability to promote degradation of Hdmx, a phosphorylation target of Chk2 and regulator of p53 activity. Besides, Chk2(S3A) failed to inhibit cell growth and, in response to IR, to arrest G(1)/S progression. These findings underscore the critical roles of S19, S33, and S35 and argue that these phosphoresidues may serve to fine-tune the ATM-dependent response of Chk2 to increasing amounts of DNA damage.


Subject(s)
Cell Cycle/physiology , DNA Damage , Protein Serine-Threonine Kinases , Serine/metabolism , 4-Nitroquinoline-1-oxide/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle/radiation effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Checkpoint Kinase 2 , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/metabolism , Humans , Hydroxyurea/metabolism , Multiprotein Complexes , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Quinolones/metabolism , RNA Interference , Radiation, Ionizing , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
15.
DNA Repair (Amst) ; 5(8): 904-13, 2006 Aug 13.
Article in English | MEDLINE | ID: mdl-16765653

ABSTRACT

The repair of DNA double-strand breaks is critical for genome integrity and tumor suppression. Here we show that following treatment with the DNA-intercalating agent actinomycin D (ActD), normal quiescent T cells accumulate double-strand breaks and die, whereas T cells from ataxia telangiectasia (AT) and Nijmegen breakage syndrome (NBS) patients are resistant to this death pathway despite a comparable amount of DNA damage. We demonstrate that the ActD-induced death pathway in quiescent T lymphocytes follows DNA damage and H2AX phosphorylation, is ATM- and NBS1-dependent and due to p53-mediated cellular apoptosis. In response to genotoxic 2-Gy gamma-irradiation, on the other hand, quiescent T cells from normal donors survive following complete resolution of the damage thus induced. T cells from AT and NBS patients also survive, but retain foci of phosphorylated H2AX due to a subtle double-strand break (DSB) repair defect. A common consequence of these two genetic defects in the DSB response is the apparent tolerance of cells containing DNA breaks. We suggest that this tolerance makes a major contribution to the oncogenic risk of patients with chromosome instability syndromes.


Subject(s)
Apoptosis/genetics , Ataxia Telangiectasia/genetics , DNA Damage , DNA Repair/genetics , Nijmegen Breakage Syndrome/genetics , T-Lymphocytes/radiation effects , DNA Primers , Dactinomycin/toxicity , Flow Cytometry , Gamma Rays , Histones/metabolism , Humans , Immunoblotting , Microscopy, Fluorescence , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
16.
Neoplasia ; 7(11): 1030-8, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16331889

ABSTRACT

Previously, it was reported that SEL1L is able to decrease the aggressive behavior of human pancreatic tumor cells both in vitro and in vivo. To gain insights into the involvement of SEL1L in tumor invasion, we performed gene expression analysis on the pancreatic cancer cell line Suit-2 subjected to two complementary strategies: upregulation and downregulation of SEL1L expression by stable transfection of the entire cDNA under an inducible promoter and by RNA-mediated interference. SuperArray and real-time analysis revealed that SEL1L modulates the expression of the matrix metalloproteinase inhibitors TIMP1 (P < .04-.03) and TIMP2 (P < .03-.05), and the PTEN gene (P < .03-.05). Gene expression modulations correlate with the decrease in invasive ability (P < .05) and in accumulation of SEL1L-expressing cells in G1. Taken together, our data indicate that SEL1L alters the expression of mediators involved in the remodeling of the extracellular matrix by creating a microenvironment that is unfavorable to invasive growth and by affecting cell cycle progression through promotion of G1 accumulation.


Subject(s)
Cell Cycle/drug effects , PTEN Phosphohydrolase/genetics , Pancreatic Neoplasms/pathology , Proteins/physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-2/genetics
17.
Hum Mol Genet ; 13(18): 2155-63, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15269180

ABSTRACT

Hypomorphic mutations of the MRE11 gene are the hallmark of the radiosensitive ataxia-telangiectasia-like disorder (ATLD). Here, we describe a new family with two affected siblings, ATLD5 and ATLD6, now aged 37 and 36, respectively. They presented with late onset cerebellar degeneration slowly progressing until puberty and absence of telangiectasias, and were cancer-free. Both patients were wild-type for ATM and NBS1, but compound heterozygotes for MRE11 gene mutations [1422C-->A, T481K; 1714C-->T, R571X]. The 1422C-->A allele was inherited from the mother, whereas the 1714C-->T, allele paternally inherited, was apparently null as a result of nonsense-mediated mRNA decay (NMD). Interestingly, the 1714C-->T mutation is the same as previously identified in an unrelated English ATLD family (probands ATLD3 and ATLD4), suggesting an important role for NMD in saving potentially lethal mutations. Lymphoblastoid cell lines (LCLs) derived from ATLD5 and ATLD6 were normal for ATM, but defective for Mre11, Rad50 and Nbs1 (the MRN complex) protein expression. Their response to gamma-radiation was abnormal, as evidenced by the enhanced radiosensitivity, attenuated autophosphorylation of ATM-S1981 and phosphorylation of the ATM targets p53-S15 and Smc1-S966, failure to form Mre11 nuclear foci and defective G1 checkpoint arrest. The fibroblasts, but not LCLs, from ATLD5 and ATLD6 showed an impaired ATM-dependent Chk2 phosphorylation. These findings further underscore the interconnection between ATM activity and MRN function, which rationalizes the clinical similarity between ataxia-telangiectasia (A-T) and ATLD.


Subject(s)
Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/metabolism , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mutation/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Adult , Alleles , Ataxia Telangiectasia/ethnology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle/genetics , Cell Nucleus/immunology , Cell Nucleus/metabolism , Cells, Cultured , Checkpoint Kinase 2 , DNA Mutational Analysis , DNA-Binding Proteins/analysis , Female , Fibroblasts/immunology , Fibroblasts/metabolism , Heterozygote , Humans , Italy , Lymphocytes/radiation effects , MRE11 Homologue Protein , Male , Nuclear Proteins/metabolism , Phosphorylation
18.
Oncogene ; 22(49): 7866-9, 2003 Oct 30.
Article in English | MEDLINE | ID: mdl-14586414

ABSTRACT

The ATM-dependent accumulation of p53 and induction of p21waf1 are key events for G1 cell-cycle checkpoint arrest following DNA damage. In ATM-null AT cells, even though the p53 and p21waf1 responses are kinetically delayed and quantitatively reduced, the G1 checkpoint is virtually disrupted, suggesting that these proteins arrive too late in G1 to enforce the arrest. As the precise mechanism remains unclear, we examined the response to DNA double-strand breaks generated by gamma-radiation (IR), to determine if ATM deficiency affects the cell-cycle phase regulation of these molecules. We find that, after irradiation, whereas normal LCL-N cells markedly increase their levels of p53 in all phases of the cell cycle, AT cells fail to show any p53 increase in the G1 phase. In addition, whereas in LCL-N p21waf1 is induced in G1 and G2-M, in AT cells this induction is partly seen in G2-M, but not in G1, indicating a different cell-cycle phase regulation of p53 and p21waf1 as a result of ATM deficiency. The levels and catalytic activity of the p53-targeting kinases ATR and DNA-PK in LCL-N and AT cells are very similar throughout the cell cycle, both before and after IR, thus excluding a phase-specific activity for these kinases. Collectively, our findings demonstrate that, in ATM-deficient cells, the p53-dependent p21waf1 response to DNA damage is not only quantitatively reduced, but also specifically suppressed in the G1 phase, thus providing a mechanistic explanation for the severe disruption of the G1 checkpoint in AT cells.


Subject(s)
Cyclins/physiology , DNA Damage , DNA-Binding Proteins , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Protein p53/physiology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/physiology , Cyclin-Dependent Kinase Inhibitor p21 , DNA-Activated Protein Kinase , G1 Phase , Humans , Nuclear Proteins , Phosphorylation , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...