Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cancer Res ; 83(17): 2824-2838, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37327406

ABSTRACT

Identifying mechanisms underlying relapse is a major clinical issue for effective cancer treatment. The emerging understanding of the importance of metastasis in hematologic malignancies suggests that it could also play a role in drug resistance and relapse in acute myeloid leukemia (AML). In a cohort of 1,273 AML patients, we uncovered that the multifunctional scavenger receptor CD36 was positively associated with extramedullary dissemination of leukemic blasts, increased risk of relapse after intensive chemotherapy, and reduced event-free and overall survival. CD36 was dispensable for lipid uptake but fostered blast migration through its binding with thrombospondin-1. CD36-expressing blasts, which were largely enriched after chemotherapy, exhibited a senescent-like phenotype while maintaining their migratory ability. In xenograft mouse models, CD36 inhibition reduced metastasis of blasts and prolonged survival of chemotherapy-treated mice. These results pave the way for the development of CD36 as an independent marker of poor prognosis in AML patients and a promising actionable target to improve the outcome of patients. SIGNIFICANCE: CD36 promotes blast migration and extramedullary disease in acute myeloid leukemia and represents a critical target that can be exploited for clinical prognosis and patient treatment.


Subject(s)
Leukemia, Myeloid, Acute , Humans , Animals , Mice , Leukemia, Myeloid, Acute/pathology , Treatment Outcome , Prognosis , Recurrence , Blast Crisis/pathology , Chronic Disease
2.
Cancers (Basel) ; 13(16)2021 Aug 12.
Article in English | MEDLINE | ID: mdl-34439212

ABSTRACT

Triple-negative breast cancer (TNBC) is notoriously aggressive with a high metastatic potential, and targeted therapies are lacking. Using transcriptomic and histologic analysis of TNBC samples, we found that a high expression of thrombospondin-1 (TSP1), a potent endogenous inhibitor of angiogenesis and an activator of latent transforming growth factor beta (TGF-ß), is associated with (i) gene signatures of epithelial-mesenchymal transition and TGF-ß signaling, (ii) metastasis and (iii) a reduced survival in TNBC patients. In contrast, in tumors expressing low levels of TSP1, gene signatures of interferon gamma (IFN-γ) signaling and lymphocyte activation were enriched. In TNBC biopsies, TSP1 expression inversely correlated with the CD8+ tumor-infiltrating lymphocytes (TILs) content. In the 4T1 metastatic mouse model of TNBC, TSP1 silencing did not affect primary tumor development but, strikingly, impaired metastasis in immunocompetent but not in immunodeficient nude mice. Moreover, TSP1 knockdown increased tumor vascularization and T lymphocyte infiltration and decreased TGF-ß activation in immunocompetent mice. Noteworthy was the finding that TSP1 knockdown increased CD8+ TILs and their programmed cell death 1 (PD-1) expression and sensitized 4T1 tumors to anti-PD-1 therapy. TSP1 inhibition might thus represent an innovative targeted approach to impair TGF-ß activation and breast cancer cell metastasis and improve lymphocyte infiltration in tumors, and immunotherapy efficacy in TNBC.

3.
Nat Cancer ; 2(11): 1204-1223, 2021 11.
Article in English | MEDLINE | ID: mdl-35122057

ABSTRACT

Therapy resistance represents a major clinical challenge in acute myeloid leukemia (AML). Here we define a 'MitoScore' signature, which identifies high mitochondrial oxidative phosphorylation in vivo and in patients with AML. Primary AML cells with cytarabine (AraC) resistance and a high MitoScore relied on mitochondrial Bcl2 and were highly sensitive to venetoclax (VEN) + AraC (but not to VEN + azacytidine). Single-cell transcriptomics of VEN + AraC-residual cell populations revealed adaptive resistance associated with changes in oxidative phosphorylation, electron transport chain complex and the TP53 pathway. Accordingly, treatment of VEN + AraC-resistant AML cells with electron transport chain complex inhibitors, pyruvate dehydrogenase inhibitors or mitochondrial ClpP protease agonists substantially delayed relapse following VEN + AraC. These findings highlight the central role of mitochondrial adaptation during AML therapy and provide a scientific rationale for alternating VEN + azacytidine with VEN + AraC in patients with a high MitoScore and to target mitochondrial metabolism to enhance the sensitivity of AML cells to currently approved therapies.


Subject(s)
Cytarabine , Leukemia, Myeloid, Acute , Azacitidine/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cytarabine/pharmacology , Humans , Leukemia, Myeloid, Acute/drug therapy , Sulfonamides
4.
World J Urol ; 33(2): 281-7, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24748552

ABSTRACT

OBJECTIVE: To evaluate the expression of CXCR4, its ligand SDF-1, ß-catenin and E-cadherin throughout the local tumor microenvironment of prostate cancer. PATIENTS AND METHODS: A total of 64 prostate cancer specimens, 24 frozen and 40 paraffin-embedded sections, were obtained from patients treated with radical prostatectomy for clinically localized cancer. Real-time RT-PCR was used for mRNA quantification of CXCR4 and SDF-1 in the tumor center (T), tumor front (F) and distant peritumoral tissue (D). Immunohistochemical analysis was used to investigate the expression patterns of CXCR4, E-cadherin and ß-catenin. Clinical records of these patients were studied for follow-up data, and the prognostic value of these molecules' expression was statistically assessed. RESULTS: CXCR4 mRNA and protein were significantly increased at the tumor front as compared to distant tissue or tumor center. In comparison, SDF-1 mRNA level gradually increased from the tumor center to the distant peritumoral tissue. High CXCR4 at the tumor front was associated with high Gleason score. Low SDF-1 at the tumor front was associated with locally advanced cancer and disease recurrence. Moreover, high CXCR4 staining at the tumor front and increased cytosolic E-cadherin expression in the same location was associated with locally advanced disease. CONCLUSIONS: CXCR4 seems overexpressed at the tumor front of prostate tumors, where it potentially promotes cell migration toward the SDF-1 centrifugal attracting gradient, as well as epithelial-mesenchymal transition. High CXCR4 and low SDF-1 levels at tumor front were both associated with adverse histological features.


Subject(s)
Biomarkers, Tumor/biosynthesis , Cadherins/biosynthesis , Chemokine CXCL12/biosynthesis , Prostatic Neoplasms/metabolism , Receptors, CXCR4/biosynthesis , beta Catenin/biosynthesis , Aged , Cell Movement , Humans , Male , Middle Aged , Prognosis , Prostatectomy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , RNA, Messenger/biosynthesis
5.
PLoS One ; 7(8): e42252, 2012.
Article in English | MEDLINE | ID: mdl-22879924

ABSTRACT

Almost all prostate cancers respond to androgen deprivation treatment but many recur. We postulated that risk of hormone escape--frequency and delay--are influenced by hormone therapy modalities. More, hormone therapies induce crucial biological changes involving androgen receptors; some might be targets for escape prevention. We investigated the relationship between the androgen deprivation treatment and the risk of recurrence using nude mice bearing the high grade, hormone-dependent human prostate cancer xenograft PAC120. Tumor-bearing mice were treated by Luteinizing-Hormone Releasing Hormone (LHRH) antagonist alone, continuous or intermittent regimen, or combined with androgen receptor (AR) antagonists (bicalutamide or flutamide). Tumor growth was monitored. Biological changes were studied as for genomic alterations, AR mutations and protein expression in a large series of recurrent tumors according to hormone therapy modalities. Therapies targeting Her-2 or AKT were tested in combination with castration. All statistical tests were two-sided. Tumor growth was inhibited by continuous administration of the LH-RH antagonist degarelix (castration), but 40% of tumors recurred. Intermittent castration or complete blockade induced by degarelix and antiandrogens combination, inhibited tumor growth but increased the risk of recurrence (RR) as compared to continuous castration (RR(intermittent): 14.5, RR(complete blockade): 6.5 and 1.35). All recurrent tumors displayed new quantitative genetic alterations and AR mutations, whatever the treatment modalities. AR amplification was found after complete blockade. Increased expression of Her-2/neu with frequent ERK/AKT activation was detected in all variants. Combination of castration with a Her-2/neu inhibitor decreased recurrence risk (0.17) and combination with an mTOR inhibitor prevented it. Anti-hormone treatments influence risk of recurrence although tumor growth inhibition was initially similar. Recurrent tumors displayed genetic instability, AR mutations, and alterations of phosphorylation pathways. We postulated that Her-2/AKT pathways allowed salvage of tumor cells under castration and we demonstrated that their inhibition prevented tumor recurrence in our model.


Subject(s)
Androgens/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays , Androgens/deficiency , Animals , Base Sequence , Castration , Cluster Analysis , Combined Modality Therapy , Disease-Free Survival , Gene Dosage/genetics , Humans , Male , Mice , Mutation/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein-Tyrosine Kinases/metabolism , Receptor, ErbB-2/genetics , Receptors, Androgen/genetics
6.
FASEB J ; 26(1): 460-7, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21982950

ABSTRACT

Markers of prostate tumor recurrence after radical prostatectomy are lacking and highly demanded. The androgen receptor (AR) is a nuclear receptor that plays a pivotal role in normal and cancerous prostate tissue. AR interacts with a number of proteins modulating its stability, localization, and activity. To test the hypothesis that an increased expression of AR partners might foster tumor development, we immunopurified AR partners in human tumors xenografted into mice. One of the identified AR partners was the multifunctional enzyme carbamoyl-phosphate synthetase II, aspartate transcarbamylase, and dihydroorotase (CAD), which catalyzes the 3 initial steps of pyrimidine biosynthesis. We combined experiments in C4-2, LNCaP, 22RV1, and PC3 human prostate cell lines and analysis of frozen radical prostatectomy samples to study the CAD-AR interaction. We show here that in prostate tumor cells, CAD fosters AR translocation into the nucleus and stimulates its transcriptional activity. Notably, in radical prostatectomy specimens, CAD expression was not correlated with proliferation markers, but a higher CAD mRNA level was associated with local tumor extension (P=0.049) and cancer relapse (P=0.017). These results demonstrate an unsuspected function for a key metabolic enzyme and identify CAD as a potential predictive marker of cancer relapse.


Subject(s)
Aspartate Carbamoyltransferase/metabolism , Biomarkers, Tumor/metabolism , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/metabolism , Dihydroorotase/metabolism , Neoplasm Recurrence, Local/diagnosis , Prostatic Neoplasms/diagnosis , Receptors, Androgen/metabolism , Androgens/metabolism , Animals , Aspartate Carbamoyltransferase/genetics , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Cytosol/metabolism , Dihydroorotase/genetics , Humans , Male , Mice , Neoplasm Recurrence, Local/metabolism , Neoplasm Transplantation , Predictive Value of Tests , Prostatic Neoplasms/metabolism , Pyrimidines/biosynthesis , RNA, Small Interfering/pharmacology , Receptors, Androgen/genetics , Transcription, Genetic/physiology , Transplantation, Heterologous
7.
Cancer Res ; 71(24): 7649-58, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22037878

ABSTRACT

The antitumor effects of pharmacologic inhibitors of angiogenesis are hampered in patients by the rapid development of tumor resistance, notably through increased invasiveness and accelerated metastasis. Here, we reevaluated the role of the endogenous antiangiogenic thrombospondin 1 (TSP1) in prostate carcinomas in which angiogenesis is an active process. In xenografted tumors, we observed that TSP1 altogether inhibited angiogenesis and fostered tumor development. Our results show that TSP1 is a potent stimulator of prostate tumor cell migration. This effect required CD36, which also mediates TSP1 antiangiogenic activity, and was mimicked by an antiangiogenic TSP1-derived peptide. As suspected for pharmacologic inhibitors of angiogenesis, the TSP1 capacities to increase hypoxia and to trigger cell migration are thus inherently linked. Importantly, although antiangiogenic TSP1 increases hypoxia in vivo, our data show that, in turn, hypoxia induced TSP1, thus generating a vicious circle in prostate tumors. In radical prostatectomy specimens, we found TSP1 expression significantly associated with invasive tumors and with tumors which eventually recurred. TSP1 may thus help select patients at risk of prostate-specific antigen relapse. Together, the data suggest that intratumor disruption of the hypoxic cycle through TSP1 silencing will limit tumor invasion.


Subject(s)
Cell Movement , Neovascularization, Pathologic/genetics , Prostatic Neoplasms/genetics , RNA Interference , Thrombospondin 1/genetics , Animals , CD36 Antigens/metabolism , Calcium/metabolism , Cell Hypoxia , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Neoplastic , Humans , Hypoxia , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Recurrence, Local , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Orchiectomy , Peptides/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , TRPV Cation Channels/metabolism , Thrombospondin 1/chemistry , Thrombospondin 1/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays/methods
8.
Cancer Res ; 70(3): 1225-35, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20103638

ABSTRACT

Castration resistance in prostate cancer (PCa) constitutes an advanced, aggressive disease with poor prognosis, associated with uncontrolled cell proliferation, resistance to apoptosis, and enhanced invasive potential. The molecular mechanisms involved in the transition of PCa to castration resistance are obscure. Here, we report that the nonselective cationic channel transient receptor potential vanilloid 2 (TRPV2) is a distinctive feature of castration-resistant PCa. TRPV2 transcript levels were higher in patients with metastatic cancer (stage M1) compared with primary solid tumors (stages T2a and T2b). Previous studies of the TRPV2 channel indicated that it is primarily involved in cancer cell migration and not in cell growth. Introducing TRPV2 into androgen-dependent LNCaP cells enhanced cell migration along with expression of invasion markers matrix metalloproteinase (MMP) 9 and cathepsin B. Consistent with the likelihood that TRPV2 may affect cancer cell aggressiveness by influencing basal intracellular calcium levels, small interfering RNA-mediated silencing of TRPV2 reduced the growth and invasive properties of PC3 prostate tumors established in nude mice xenografts, and diminished expression of invasive enzymes MMP2, MMP9, and cathepsin B. Our findings establish a role for TRPV2 in PCa progression to the aggressive castration-resistant stage, prompting evaluation of TRPV2 as a potential prognostic marker and therapeutic target in the setting of advanced PCa.


Subject(s)
Prostatic Neoplasms/genetics , RNA Interference , TRPV Cation Channels/genetics , Androgens/metabolism , Androgens/pharmacology , Animals , Blotting, Western , Cell Line, Tumor , Cell Movement , Disease Progression , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Male , Mice , Mice, Nude , Microscopy, Confocal , Neoplasm Invasiveness , Neoplasm Metastasis , Orchiectomy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Reverse Transcriptase Polymerase Chain Reaction , TRPV Cation Channels/metabolism , TRPV Cation Channels/physiology , Xenograft Model Antitumor Assays
9.
Curr Top Med Chem ; 9(12): 1117-29, 2009.
Article in English | MEDLINE | ID: mdl-19860712

ABSTRACT

Synthetic small interfering RNAs (siRNAs) open promising new therapeutic perspectives in acute and chronic pathologies. A number of experiments in mice demonstrated the ability of naked siRNAs injected under a normal pressure to trigger gene silencing in vivo, translating into a measurable phenotype. We focus in this review on the information that we can gain from these experiments, and discuss how the specificity of the gene silencing in vivo can be controlled. Because the activity of most drugs increases with the dosing, we are prone to consider that increasing the concentration of siRNAs within cells enhances the efficiency and the duration of the silencing. However, because RNAi is a saturable process, and because increasing the siRNA concentration into cells can induce undesirable side effects, this must be demonstrated. We compare in this review the methods used to quantify and study the biodistribution of siRNAs in living animals, and discuss how these methods can help in designing for each model and each siRNA the most adequate protocol to silence a cognate target gene in vivo.


Subject(s)
Drug Delivery Systems , RNA, Small Interfering , Animals , Gene Silencing , Mice
10.
J Natl Cancer Inst ; 101(4): 237-47, 2009 Feb 18.
Article in English | MEDLINE | ID: mdl-19211441

ABSTRACT

BACKGROUND: Netrin-1 may promote colorectal and breast tumorigenesis, by inhibiting apoptosis induced by its dependence receptors, deleted in colorectal cancer (DCC) and uncoordinated-5-homolog (UNC5H). The status of netrin-1 and its receptors in non-small cell lung cancer (NSCLC) was unknown. METHODS: The levels of netrin-1 and its receptors were analyzed in a panel of 92 NSCLC and 25 human lung cancer cell lines by quantitative reverse transcription-polymerase chain reaction and immunohistochemistry. In lung cancer cell lines that express netrin-1, the expression of netrin-1 was inhibited by using small interfering RNA (siRNA), or interference with netrin-1 was performed by treatment with a decoy recombinant DCC ectodomain protein (DCC-5Fbn). Cell death was monitored with a trypan blue exclusion assay or by measuring caspase-3 activity. The effect of netrin-1 interference on tumor growth was analyzed by DCC-5Fbn intratumoral or netrin-1 siRNA intraperitoneal injection in mice engrafted with lung cancer cell lines. All statistical tests were two-sided. RESULTS: High levels of netrin-1 were found in 43 of the 92 NSCLC tumor samples (47%). Interference with netrin-1 in human lung cancer cell lines was associated with UNC5H-mediated cell death in vitro (percentage of cell death in untreated and in DCC-5Fbn-treated cells = 8% and 26%, respectively, difference = 18%, 95% confidence interval [CI] = 10% to 26%; P = .049) and with lung tumor growth inhibition and/or regression in xenografted nude mice (12 mice in DCC-5Fbn-treated group and 13 mice in control group). Mean volume of control and DCC-5Fbn-treated tumors on day 46 was 489 and 84 mm(3), respectively (difference = 404 mm(3), 95% CI = 145 to 664 mm(3); P < .001). CONCLUSIONS: Almost half of the NSCLC tissue samples examined expressed high levels of netrin-1. Extracellular targeting of the interaction between netrin-1 and UNC5H may be a promising therapeutic approach for NSCLCs that express netrin-1.


Subject(s)
Apoptosis , Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/chemistry , Lung Neoplasms/chemistry , Nerve Growth Factors/analysis , RNA, Small Interfering/analysis , Receptors, Cell Surface/analysis , Tumor Suppressor Proteins/analysis , Animals , Apoptosis Regulatory Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Coloring Agents , DCC Receptor , Death-Associated Protein Kinases , Disease Progression , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Nude , Nerve Growth Factors/genetics , Netrin Receptors , Netrin-1 , Plasmids , Receptors, Cell Surface/genetics , Recombinant Proteins , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Transplantation, Heterologous , Trypan Blue , Tumor Suppressor Proteins/genetics
11.
PLoS One ; 2(10): e1006, 2007 Oct 10.
Article in English | MEDLINE | ID: mdl-17925854

ABSTRACT

BACKGROUND: Prostate carcinomas are initially dependent on androgens, and castration or androgen antagonists inhibit their growth. After some time though, tumors become resistant and recur with a poor prognosis. The majority of resistant tumors still expresses a functional androgen receptor (AR), frequently amplified or mutated. METHODOLOGY/PRINCIPAL FINDINGS: To test the hypothesis that AR is not only expressed, but is still a key therapeutic target in advanced carcinomas, we injected siRNA targeting AR into mice bearing exponentially growing castration-resistant tumors. Quantification of siRNA into tumors and mouse tissues demonstrated their efficient uptake. This uptake silenced AR in the prostate, testes and tumors. AR silencing in tumors strongly inhibited their growth, and importantly, also markedly repressed the VEGF production and angiogenesis. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that carcinomas resistant to hormonal manipulations still depend on the expression of the androgen receptor for their development in vivo. The siRNA-directed silencing of AR, which allows targeting overexpressed as well as mutated isoforms, triggers a strong antitumoral and antiangiogenic effect. siRNA-directed silencing of this key gene in advanced and resistant prostate tumors opens promising new therapeutic perspectives and tools.


Subject(s)
Carcinoma/genetics , Carcinoma/pathology , Gene Silencing , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Small Interfering/metabolism , Receptors, Androgen/genetics , Animals , Cell Line, Tumor , Humans , Male , Mice , Neoplasm Transplantation , Neovascularization, Pathologic , Prostate/metabolism , Testis/metabolism , Vascular Endothelial Growth Factor A/metabolism
12.
FASEB J ; 20(3): 476-8, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16423876

ABSTRACT

Spermatogenesis is a complex process involving cell proliferation, differentiation, and apoptosis. Fibroblast growth factor 2 (FGF-2) is involved in testicular function, but its role in spermatogenesis has not been fully documented. The control of FGF-2 expression particularly occurs at the translational level, by an internal ribosome entry site (IRES)-dependent mechanism driving the use of alternative initiation codons. To study IRES activity regulation in vivo, we have developed transgenic mice expressing a bicistronic construct coding for two luciferase genes. Here, we show that the FGF-2 IRES is age-dependently activated in mouse testis, whereas EMCV and c-myc IRESs are not. Real-time PCR confirms that this regulation is translational. By using immunohistological techniques, we demonstrate that FGF-2 IRES stimulation occurs in adult, but not in immature, type-A spermatogonias. This is correlated with activation of endogenous FGF-2 expression in spermatogonia; whereas FGF-2 mRNA transcription is known to decrease in adult testis. Interestingly, the FGF-2 IRES activation is triggered by testosterone and is partially inhibited by siRNA directed against the androgen receptor. Two-dimensional analysis of proteins bound to the FGF-2 mRNA 5'UTR after UV cross-linking reveals that testosterone treatment correlates with the binding of several proteins. These data suggest a paracrine loop where IRES-dependent FGF-2 expression, stimulated by Sertoli cells in response to testosterone produced by Leydig cells, would in turn activate Leydig function and testosterone production. In addition, nuclear FGF-2 isoforms could be involved in an intracrine function of FGF-2 in the start of spermatogenesis, mitosis, or meiosis initiation. This report demonstrates that mRNA translation regulation by an IRES-dependent mechanism participates in a physiological process.


Subject(s)
Fibroblast Growth Factor 2/physiology , Leydig Cells/physiology , Protein Biosynthesis , RNA, Messenger/genetics , Regulatory Sequences, Nucleic Acid , Sertoli Cells/physiology , Spermatogenesis/physiology , Testis/physiology , Testosterone/physiology , 5' Untranslated Regions , Age Factors , Androgen Receptor Antagonists , Animals , Codon , Fibroblast Growth Factor 2/biosynthesis , Fibroblast Growth Factor 2/genetics , Genes, Reporter , Genes, Synthetic , Luciferases, Renilla/genetics , Male , Meiosis , Mice , Mice, Transgenic , Mitosis , Paracrine Communication , Peptide Chain Initiation, Translational/physiology , Protein Isoforms/physiology , RNA, Messenger/radiation effects , RNA, Small Interfering/pharmacology , Receptors, Androgen/genetics , Recombinant Fusion Proteins/physiology , Ribosomes/metabolism , Testis/growth & development , Testis/metabolism , Testosterone/metabolism , Testosterone/pharmacology , Ultraviolet Rays
13.
Int J Cancer ; 116(5): 686-91, 2005 Sep 20.
Article in English | MEDLINE | ID: mdl-15838828

ABSTRACT

The antiangiogenic extracellular matrix protein thrombospondin-1 (TSP-1) inhibits tumor growth and metastasis in animals. However, the clinical relevance of such findings are equivocal as increased stromal TSP-1 expression has been associated with either good or poor prognosis. In an effort to obtain a more integrated understanding of the role of TSP-1 in breast cancer, we first used a breast tumorigenesis model in which tumor-associated stromal fibroblasts were engineered to produce high levels of TSP-1. We demonstrate here that stromal TSP-1 delayed human MDA-MB-231/B02 breast tumor growth. However, this delay in MDA-MB-231/B02 tumor growth upon exposure to TSP-1 was associated with an increased vascular endothelial growth factor (VEGF) expression in tumor cells themselves, leading to a tumor growth rate comparable to that of tumors whose fibroblasts did not overproduce TSP-1. Clinical evidence also suggested that primary breast carcinomas have adapted to escape the effects of stromal TSP-1. TSP-1 was found to be expressed in the stroma of human breast carcinomas where, although its level correlated with decreased vascularization, it was unexpectedly associated with a reduction of relapse-free survival. In metastatic axillary lymph nodes, tumor cells expressed high levels of VEGF and TSP-1 expression were no longer associated with a decreased vascularization. Overall, these results suggest that a resistance may develop early in human breast cancers as a result of high in situ exposure to stromal TSP-1, leading to disease progression.


Subject(s)
Breast Neoplasms/blood supply , Neovascularization, Pathologic/prevention & control , Thrombospondin 1/physiology , Animals , Breast Neoplasms/pathology , Cell Movement/drug effects , Disease Progression , Female , Humans , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasm Transplantation , Thrombospondin 1/analysis , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/analysis
14.
Cancer Res ; 65(1): 300-8, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15665307

ABSTRACT

In order to understand why the angiogenesis inhibitor thrombospondin-1 (TSP1) is often, although not always, associated with prostatic tumors, we have investigated its relationship with the testosterone and the vasculature on which both normal and tumorigenic prostatic epithelia depend. In vivo, androgen withdrawal led to increased TSP1 production and decreased vascularization in the normal rat prostate which was reversed by androgen replacement. Androgen repression of TSP1 production occurred at the transcriptional level and was dependent on the presence of the first intron of the TSP1 gene. In an experimental model of prostate tumorigenesis, TSP1, when delivered by admixed stromal fibroblasts, markedly delayed LNCaP tumor growth and limited tumor vascularization. However, prolonged exposure to TSP1 resulted in the growth of tumors secreting high levels of vascular endothelial growth factor in the bloodstream of tumor-bearing animals and tumor growth was no longer sensitive to TSP1 inhibitory effects. Clinical evidence also suggested that prostate carcinomas are able to adapt to escape the antiangiogenic effects of TSP1. In human androgen-dependent localized prostate carcinomas, TSP1 expression was inversely correlated with blood vessel density. Androgen deprivation in patients with hormone-responsive tumors led to increased TSP1 expression and vascular regression. In contrast, despite a sustained expression in the tumor bed, TSP1 was no longer associated with decreased vascularization in hormone-refractory prostate tumors. Overall, these results suggest that the high in situ TSP1 exposure triggered by androgen deprivation in patients with prostate cancer could lead to early tumor resistance. Such patients could benefit from a combination of androgen deprivation and antiangiogenic therapy in order to minimize the induction of such tumor escape.


Subject(s)
Androgens/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation/drug effects , Neovascularization, Pathologic/prevention & control , Neovascularization, Physiologic/drug effects , Prostatic Neoplasms/blood supply , Thrombospondin 1/genetics , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Humans , Male , Mice , Mice, Nude , Orchiectomy , Prostate , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Rats , Rats, Sprague-Dawley , Thrombospondin 1/drug effects , Vascular Endothelial Growth Factor A/analysis
15.
EMBO J ; 23(3): 605-15, 2004 Feb 11.
Article in English | MEDLINE | ID: mdl-14765126

ABSTRACT

The Rb/E2F complex represses S-phase genes both in cycling cells and in cells that have permanently exited from the cell cycle and entered a terminal differentiation pathway. Here we show that S-phase gene repression, which involves histone-modifying enzymes, occurs through distinct mechanisms in these two situations. We used chromatin immunoprecipitation to show that methylation of histone H3 lysine 9 (H3K9) occurs at several Rb/E2F target promoters in differentiating cells but not in cycling cells. Furthermore, phenotypic knock-down experiments using siRNAs showed that the histone methyltransferase Suv39h is required for histone H3K9 methylation and subsequent repression of S-phase gene promoters in differentiating cells, but not in cycling cells. These results indicate that the E2F target gene permanent silencing mechanism that is triggered upon terminal differentiation is distinct from the transient repression mechanism in cycling cells. Finally, Suv39h-depleted myoblasts were unable to express early or late muscle differentiation markers. Thus, appropriately timed H3K9 methylation by Suv39h seems to be part of the control switch for exiting the cell cycle and entering differentiation.


Subject(s)
Cell Differentiation/physiology , Gene Silencing/physiology , Histones/metabolism , Methyltransferases/metabolism , Repressor Proteins/metabolism , S Phase/physiology , Animals , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , E2F Transcription Factors , HeLa Cells , Humans , Mice , Myoblasts/physiology , NIH 3T3 Cells , Promoter Regions, Genetic/physiology , RNA, Small Interfering , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism
16.
Cancer Res ; 63(14): 3919-22, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12873985

ABSTRACT

In the past few years, several laboratories have developed antiangiogenic molecules that starve tumors by targeting their vasculature and we have shown that, when produced in tumors, the antiangiogenic molecule thrombospondin-1 (TSP1) reduces the vascularization and delays tumor onset. Yet over time, tumor cells producing active TSP1 do eventually form exponentially growing tumors. These tumors are composed of cells secreting unusually high amounts of the angiogenic stimulator vascular endothelial growth factor (VEGF) that are sufficient to overcome the inhibitory TSP1. Here, we use short double-stranded RNA (siRNA) to trigger RNA interference and thereby impair the synthesis of VEGF and ask if this inability to produce VEGF prevents the development of TSP1 resistance. Systemic in vivo administration of crude anti-VEGF siRNA reduced the growth of unaltered fibrosarcoma tumor cells, and when the anti-VEGF siRNA was expressed from tumor cells themselves, such inhibition was synergistic with the inhibitory effects derived from TSP1 secretion by the tumor cells. Anti-VEGF siRNA delayed the emergence of TSP1-resistant tumors and strikingly reduced their subsequent growth rate.


Subject(s)
Endothelial Growth Factors/antagonists & inhibitors , Fibrosarcoma/blood supply , Lymphokines/antagonists & inhibitors , Neovascularization, Pathologic/prevention & control , RNA, Small Interfering/genetics , Thrombospondin 1/physiology , Animals , Cell Division/genetics , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Female , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Lymphokines/biosynthesis , Lymphokines/genetics , Mice , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Rats , Thrombospondin 1/biosynthesis , Thrombospondin 1/genetics , Transfection , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
17.
Cancer Res ; 62(22): 6538-44, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12438248

ABSTRACT

Bisphosphonates (BPs) are used currently in the treatment of patients with bone metastases because these compounds inhibit bone resorption. We examined here the effects of BPs on inhibition of endothelial cell functions in vitro and in vivo. Treatment of endothelial cells with BPs (clodronate, risedronate, ibandronate, and zoledronic acid) reduced proliferation, induced apoptosis, and decreased capillary-like tube formation in vitro. Quantification of blood vessels in bone biopsy specimens from patients with Paget's disease before and after clodronate treatment showed a 40% reduction of the vascularization after BP treatment. However, such a decreased vascularity could be secondary to a reduction of bone resorption. Therefore, the tissue distribution of [14C]BPs in male rats was examined to develop an angiogenesis model in a noncalcified tissue where BPs could accumulate. [14C]BPs (zoledronic acid, ibandronate, and clodronate) not only accumulated in bone but also transiently accumulated in the prostate. The effects of BPs on testosterone-induced revascularization of the prostate gland in castrated rats were then studied. Testosterone in combination with ibandronate or zoledronic acid induced a 17-35% reduction of the prostate weight compared with castrated rats treated with testosterone alone. Blood vessel immunostaining on prostate tissue sections revealed that both ibandronate and zoledronic acid induced a 50% reduction of the revascularization of the prostate gland. Moreover, zoledronic acid did not alter testosterone-induced activity of a luciferase gene reporter construct transfected in androgen-dependent prostatic cells, indicating that this BP did not directly interfere with testosterone. In conclusion, BPs have in vivo antiangiogenic properties, which could be of relevance to improve therapy and prevention of bone metastasis. In addition, our results extend the potential clinical use of BPs to patients with early prostate cancer.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Diphosphonates/pharmacology , Etidronic Acid/analogs & derivatives , Neovascularization, Physiologic/drug effects , Prostate/blood supply , Testosterone/pharmacology , Angiogenesis Inhibitors/pharmacokinetics , Animals , Bone and Bones/blood supply , Bone and Bones/pathology , Cell Division/drug effects , Cells, Cultured , Clodronic Acid/pharmacokinetics , Clodronic Acid/pharmacology , Diphosphonates/pharmacokinetics , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Etidronic Acid/pharmacokinetics , Etidronic Acid/pharmacology , Humans , Ibandronic Acid , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Male , Orchiectomy , Osteitis Deformans/pathology , Prostate/cytology , Prostate/drug effects , Rats , Rats, Sprague-Dawley , Risedronic Acid , Testosterone/antagonists & inhibitors , Tissue Distribution , Zoledronic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...