Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Sci Transl Med ; 12(531)2020 02 19.
Article in English | MEDLINE | ID: mdl-32075943

ABSTRACT

Topoisomerase I (TOP1) inhibitors trap TOP1 cleavage complexes resulting in DNA double-strand breaks (DSBs) during replication, which are repaired by homologous recombination (HR). Triple-negative breast cancer (TNBC) could be eligible for TOP1 inhibitors given the considerable proportion of tumors with a defect in HR-mediated repair (BRCAness). The TOP1 inhibitor irinotecan was tested in 40 patient-derived xenografts (PDXs) of TNBC. BRCAness was determined with a single-nucleotide polymorphism (SNP) assay, and expression of Schlafen family member 11 (SLFN11) and retinoblastoma transcriptional corepressor 1 (RB1) was evaluated by real-time polymerase chain reaction (RT-PCR) and immunohistochemistry analyses. In addition, the combination of irinotecan and the ataxia telangiectasia and Rad3-related protein (ATR) inhibitor VE-822 was tested in SLFN11-negative PDXs, and two clinical non-camptothecin TOP1 inhibitors (LMP400 and LMP776) were tested. Thirty-eight percent of the TNBC models responded to irinotecan. BRCAness combined with high SLFN11 expression and RB1 loss identified highly sensitive tumors, consistent with the notion that deficiencies in cell cycle checkpoints and DNA repair result in high sensitivity to TOP1 inhibitors. Treatment by the ATR inhibitor VE-822 increased sensitivity to irinotecan in SLFN11-negative PDXs and abolished irinotecan-induced phosphorylation of checkpoint kinase 1 (CHK1). LMP400 (indotecan) and LMP776 (indimitecan) showed high antitumor activity in BRCA1-mutated or BRCAness-positive PDXs. Last, low SLFN11 expression was associated with poor survival in 250 patients with TNBC treated with anthracycline-based chemotherapy. In conclusion, a substantial proportion of TNBC respond to irinotecan. BRCAness, high SLFN11 expression, and RB1 loss are highly predictive of response to irinotecan and the clinical indenoisoquinoline TOP1 inhibitors.


Subject(s)
Topoisomerase I Inhibitors , Triple Negative Breast Neoplasms , Humans , Irinotecan/pharmacology , Irinotecan/therapeutic use , Nuclear Proteins/metabolism , Retinoblastoma Binding Proteins , Topoisomerase I Inhibitors/pharmacology , Topoisomerase I Inhibitors/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Ubiquitin-Protein Ligases
2.
Int J Cancer ; 145(7): 1902-1912, 2019 10 01.
Article in English | MEDLINE | ID: mdl-30859564

ABSTRACT

Triple-negative breast cancer (TNBC) represents 10% of all breast cancers and is a very heterogeneous disease. Globally, women with TNBC have a poor prognosis, and the development of effective targeted therapies remains a real challenge. Patient-derived xenografts (PDX) are clinically relevant models that have emerged as important tools for the analysis of drug activity and predictive biomarker discovery. The purpose of this work was to analyze the molecular heterogeneity of a large panel of TNBC PDX (n = 61) in order to test targeted therapies and identify biomarkers of response. At the gene expression level, TNBC PDX represent all of the various TNBC subtypes identified by the Lehmann classification except for immunomodulatory subtype, which is underrepresented in PDX. NGS and copy number data showed a similar diversity of significantly mutated gene and somatic copy number alteration in PDX and the Cancer Genome Atlas TNBC patients. The genes most commonly altered were TP53 and oncogenes and tumor suppressors of the PI3K/AKT/mTOR and MAPK pathways. PDX showed similar morphology and immunohistochemistry markers to those of the original tumors. Efficacy experiments with PI3K and MAPK inhibitor monotherapy or combination therapy showed an antitumor activity in PDX carrying genomic mutations of PIK3CA and NRAS genes. TNBC PDX reproduce the molecular heterogeneity of TNBC patients. This large collection of PDX is a clinically relevant platform for drug testing, biomarker discovery and translational research.


Subject(s)
Gene Dosage , Gene Expression Profiling/methods , Gene Regulatory Networks , High-Throughput Nucleotide Sequencing/methods , Triple Negative Breast Neoplasms/genetics , Animals , Class I Phosphatidylinositol 3-Kinases/genetics , Female , GTP Phosphohydrolases/genetics , Gene Expression Regulation, Neoplastic , Genetic Heterogeneity , Humans , Membrane Proteins/genetics , Mice , Middle Aged , Molecular Targeted Therapy , Neoplasm Transplantation , Precision Medicine , Signal Transduction , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Tumor Suppressor Protein p53/genetics
3.
J Vet Diagn Invest ; 31(1): 142-145, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30541399

ABSTRACT

An 8-y-old, intact female degu ( Octodon degus) was presented with a slow-growing mass on the tail tip. The mass was completely removed by partial caudectomy. Histologically, the last coccygeal vertebra was replaced by a lobulated neoplasm composed of large clear polygonal cells embedded in a myxoid alcian blue-positive matrix with highly vacuolated cytoplasm (physaliferous cells) and intracytoplasmic periodic acid-Schiff-positive granules. The neoplasm exhibited the morphologic features of a "classic" chordoma of humans, which is 1 of 3 distinct chordoma subtypes. Immunohistochemistry revealed dual expression of cytokeratin AE1/AE3 and vimentin, consistent with a diagnosis of chordoma. Chordomas are uncommon slow-growing neoplasms in humans and animals, arising from notochordal remnants. Depending on their subtype and location, they can have a high local recurrence rate and metastatic risk. Chordoma should be included in the differential diagnosis of a soft tissue mass on the tail of a degu, similar to the clinical situation in ferrets.


Subject(s)
Chordoma/veterinary , Octodon , Animals , Chordoma/diagnosis , Chordoma/etiology , Chordoma/pathology , Diagnosis, Differential , Female , Immunohistochemistry/veterinary , Sacrococcygeal Region/pathology
4.
Oncotarget ; 9(51): 29587-29600, 2018 Jul 03.
Article in English | MEDLINE | ID: mdl-30038706

ABSTRACT

Breast cancer is a complex disease in which each patient could present several genetic alterations that are therapeutically relevant in cancers. Here we explored the therapeutic benefit of combining PARP and mTOR inhibitors in a context of DNA repair deficiency and PI3K pathway activation. The combination of everolimus and olaparib was tested in BRCA2-mutated patient-derived xenografts (PDX) carrying alterations in the PI3K/AKT/mTOR pathway. An RPPA analysis of different signalling pathways was performed in untreated and treated xenografts. Everolimus and olaparib showed marked anti-tumor activities in the monotherapy setting and high efficacy when given in combination with 100% of mice showing tumor regressions. The fraction of P-H2AX positive cells was increased in both monotherapy arms and strongly increased in the combination setting. Everolimus given as monotherapy resulted in downregulation of different proteins involved in DNA damage repair, including FANCD2, RAD50 and SUV39H1. In the combination setting, expression of these proteins was almost completely abolished, suggesting convergence of PARP and mTOR in downregulation of DNA damage repair components. In conclusion, our results suggest that combining mTOR and DNA repair inhibition could be a successful strategy to treat a subset of breast cancer with BRCA2 mutation and alterations in the PI3K/AKT/mTOR pathway.

5.
J Pathol ; 246(1): 103-114, 2018 09.
Article in English | MEDLINE | ID: mdl-29876931

ABSTRACT

To ensure their high proliferation rate, tumor cells have an iron metabolic disorder causing them to have increased iron needs, making them more susceptible to iron deprivation. This vulnerability could be a therapeutic target. In breast cancers, the development of new therapeutic approaches is urgently needed for patients with triple-negative tumors, which frequently relapse after chemotherapy and suffer from a lack of targeted therapies. In this study, we demonstrated that deferasirox (DFX) synergises with standard chemotherapeutic agents such as doxorubicin, cisplatin and carboplatin to inhibit cell proliferation and induce apoptosis and autophagy in triple-negative breast cancer (TNBC) cells. Moreover, the combination of DFX with doxorubicin and cyclophosphamide delayed recurrences in breast cancer patient-derived xenografts without increasing the side-effects of chemotherapies alone or altering the global iron storage of mice. Antitumor synergy of DFX and doxorubicin seems to involve downregulation of the phosphoinositide 3-kinase and nuclear factor-κB pathways. Iron deprivation in combination with chemotherapy could thus help to improve the effectiveness of chemotherapy in TNBC patients without increasing toxicity. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carboplatin/pharmacology , Cisplatin/pharmacology , Deferasirox/pharmacology , Doxorubicin/pharmacology , Iron Chelating Agents/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Proliferation/drug effects , Drug Synergism , Humans , Iron/metabolism , MCF-7 Cells , Mice, Nude , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Signal Transduction/drug effects , Time Factors , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
6.
Clin Cancer Res ; 24(11): 2605-2615, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29463559

ABSTRACT

Purpose: Triple-negative breast cancer (TNBC) patients with residual disease after neoadjuvant chemotherapy have a poor outcome. We developed patient-derived xenografts (PDX) from residual tumors to identify efficient chemotherapies and predictive biomarkers in a context of resistance to anthracyclines- and taxanes-based treatments.Experimental Design: PDX were established from residual tumors of primary breast cancer patients treated in neoadjuvant setting. TNBC PDX were treated by anthracyclines, taxanes, platins, and capecitabine. Predictive biomarkers were identified by transcriptomic and immunohistologic analysis. Downregulation of RB1 was performed by siRNA in a cell line established from a PDX.Results: Residual TNBC PDX were characterized by a high tumor take, a short latency, and a poor prognosis of the corresponding patients. With the exception of BRCA1/2-mutated models, residual PDX were resistant to anthracyclines, taxanes, and platins. Capecitabine, the oral prodrug of 5-FU, was highly efficient in 60% of PDX, with two models showing complete responses. Prior treatment of a responder PDX with 5-FU increased expression of thymidylate synthase and decreased efficacy of capecitabine. Transcriptomic and IHC analyses of 32 TNBC PDX, including both residual tumors and treatment-naïve derived tumors, identified RB1 and TYMP proteins as predictive biomarkers for capecitabine response. Finally, RB1 knockdown in a cell line established from a capecitabine-responder PDX decreased sensitivity to 5-FU treatment.Conclusions: We identified capecitabine as efficient chemotherapy in TNBC PDX models established from residual disease and resistant to anthracyclines, taxanes, and platins. RB1 positivity and high expression of TYMP were significantly associated with capecitabine response. Clin Cancer Res; 24(11); 2605-15. ©2018 AACR.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Capecitabine/pharmacology , Retinoblastoma Binding Proteins/genetics , Thymidine Phosphorylase/genetics , Triple Negative Breast Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Antimetabolites, Antineoplastic/therapeutic use , Capecitabine/therapeutic use , Cell Proliferation , Disease Models, Animal , Drug Resistance, Neoplasm , Female , Fluorouracil/pharmacology , Gene Expression Profiling , Gene Silencing , Humans , Mice , RNA, Small Interfering/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
7.
J Feline Med Surg ; 19(2): 85-93, 2017 02.
Article in English | MEDLINE | ID: mdl-26316516

ABSTRACT

Objectives The aim of the study was to describe the ultrasonographic and endoscopic appearance and characteristics of the caecum in asymptomatic cats, and to correlate these findings with histology. Methods Ex vivo ultrasonographic and histologic evaluations of a fresh caecum were initially performed. Then, 20 asymptomatic cats, privately owned or originating from a reproductive colony, were recruited. All cats had an ultrasonographic examination of the ileocaecocolic junction, where the thickness of the caecal wall, ileocolic lymph nodes and the echogenicity of the local fat were assessed. They all underwent a colonoscopy with a macroscopic assessment of the mucosa and biopsies for histology. Results An ultrasonographic hypoechoic nodular inner layer, which corresponded to the coalescence of multiple lymphoid follicles originating from the submucosa and protruding in the mucosa on histology, was visible in all parts of the caecum. The combined mucosa and submucosa was measured ultrasonographically and defined as the follicular layer. Although all cats were asymptomatic, 3/19 cats showed mild caecal inflammation on histology. The most discriminatory ultrasonographic parameter in assessing this subclinical inflammation was the thickness of the follicular layer at the entrance of the caecum, with a cut-off value of 2.0 mm. All cats (20/20) showed some degree of macroscopic 'dimpling' of the caecal mucosa on endoscopy. Conclusions and relevance Lymphoid follicles in the caecal mucosa and submucosa constitute a unique follicular layer on ultrasound. In asymptomatic cats, a subtle, non-clinically relevant inflammation may exist and this is correlated with an increased thickness of the follicular layer on ultrasound. On endoscopy, a 'dimpled aspect' to the caecal mucosa is a normal finding in the asymptomatic cat.


Subject(s)
Cats/anatomy & histology , Cecum/anatomy & histology , Animals , Biopsy/veterinary , Cecum/diagnostic imaging , Colonoscopy/veterinary , Female , Male , Prospective Studies , Reference Values , Ultrasonography/veterinary
8.
Oncotarget ; 7(30): 48206-48219, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27374081

ABSTRACT

Triple-negative breast cancers (TNBC) are characterized by frequent alterations in the PI3K/AKT/mTOR signaling pathway. In this study, we analyzed PI3K pathway activation in 67 patient-derived xenografts (PDX) of breast cancer and investigated the anti-tumor activity of the mTOR inhibitor everolimus in 15 TNBC PDX with different expression and mutational status of PI3K pathway markers. Expression of the tumor suppressors PTEN and INPP4B was lost in 55% and 76% of TNBC PDX, respectively, while mutations in PIK3CA and AKT1 genes were rare. In 7 PDX treatment with everolimus resulted in a tumor growth inhibition higher than 50%, while 8 models were classified as low responder or resistant. Basal-like, LAR (Luminal AR), mesenchymal and HER2-enriched tumors were present in both responder and resistant groups, suggesting that tumor response to everolimus is not restricted to a specific TNBC subtype. Analysis of treated tumors showed a correlation between tumor response and post-treatment phosphorylation of AKT, increased in responder PDX, while PI3K pathway markers at baseline were not sufficient to predict everolimus response. In conclusion, targeting mTOR decreased tumor growth in 7 out of 15 TNBC PDX tested. Response to everolimus occurred in different TNBC subtypes and was associated with post-treatment increase of P-AKT.


Subject(s)
Everolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Animals , Cell Growth Processes/drug effects , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Mice , Mice, Nude , Molecular Targeted Therapy , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
9.
Br J Cancer ; 114(2): 177-87, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26695443

ABSTRACT

BACKGROUND: Oestrogen receptor-negative (ER-) breast cancer is intrinsically sensitive to chemotherapy. However, tumour response is often incomplete, and relapse occurs with high frequency. The aim of this work was to analyse the molecular characteristics of residual tumours and early response to chemotherapy in patient-derived xenografts (PDXs) of breast cancer. METHODS: Gene and protein expression profiles were analysed in a panel of ER- breast cancer PDXs before and after chemotherapy treatment. Tumour and stromal interferon-gamma expression was measured in xenografts lysates by human and mouse cytokine arrays, respectively. RESULTS: The analysis of residual tumour cells in chemo-responder PDX revealed a strong overexpression of IFN-inducible genes, induced early after AC treatment and associated with increased STAT1 phosphorylation, DNA-damage and apoptosis. No increase in IFN-inducible gene expression was observed in chemo-resistant PDXs upon chemotherapy. Overexpression of IFN-related genes was associated with human IFN-γ secretion by tumour cells. CONCLUSIONS: Treatment-induced activation of the IFN/STAT1 pathway in tumour cells is associated with chemotherapy response in ER- breast cancer. Further validations in prospective clinical trials will aim to evaluate the usefulness of this signature to assist therapeutic strategies in the clinical setting.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Interferon-gamma/drug effects , Receptors, Estrogen/metabolism , STAT1 Transcription Factor/drug effects , Adaptor Proteins, Signal Transducing , Animals , Antigens/drug effects , Antigens/genetics , Antigens/metabolism , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Capecitabine/pharmacology , Carrier Proteins/drug effects , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 3/drug effects , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/drug effects , Caspase 7/genetics , Caspase 7/metabolism , Cisplatin/pharmacology , Cytokines/drug effects , Cytokines/genetics , Cytokines/metabolism , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Female , Gene Expression Profiling , Humans , Immunohistochemistry , In Situ Hybridization , Interferon-beta/drug effects , Interferon-beta/genetics , Interferon-beta/metabolism , Interferon-gamma/genetics , Interferon-gamma/metabolism , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Nude , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Myxovirus Resistance Proteins/drug effects , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/metabolism , Neoplasm Transplantation
10.
Int J Cancer ; 138(10): 2510-21, 2016 May 15.
Article in English | MEDLINE | ID: mdl-26686064

ABSTRACT

The receptor tyrosine kinase RET is implicated in the progression of luminal breast cancers (BC) but its role in estrogen receptor (ER) negative tumors is unknown. Here we investigated the expression of RET in breast cancer patients tumors and patient-derived xenografts (PDX) and evaluated the therapeutic potential of Vandetanib, a tyrosin kinase inhibitor with strong activity against RET, EGFR and VEGFR2, in ER negative breast cancer PDX. The RT-PCR analysis of RET expression in breast tumors of 446 patients and 57 PDX, showed elevated levels of RET in ER+ and HER2+ subtypes and in a small subgroup of triple-negative breast cancers (TNBC). The activity of Vandetanib was tested in vivo in three PDX models of TNBC and one model of HER2+ BC with different expression levels of RET and EGFR. Vandetanib induced tumor regression in PDX models with high expression of RET or EGFR. The effect was associated with inhibition of RET/EGFR phosphorylation and MAP kinase pathway and increased necrosis. In a PDX model with no expression of RET nor EGFR, Vandetanib slowed tumor growth without inducing tumor regression. In addition, treatment by Vandetanib decreased expression of murine Vegf receptors and the endothelial marker Cd31 in the four PDX models tested, suggesting inhibition of tumor vascularization. In summary, these preclinical results suggest that Vandetanib treatment could be useful for patients with ER negative breast cancers overexpressing Vandetanib's main targets.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Receptors, Estrogen/deficiency , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Disease Models, Animal , Female , Gene Expression , Humans , MAP Kinase Signaling System/drug effects , Mice , Middle Aged , Molecular Targeted Therapy , Neoplasm Grading , Neoplasm Metastasis , Neovascularization, Pathologic/drug therapy , Phosphorylation/drug effects , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Quinazolines/therapeutic use , RNA, Messenger/genetics , Receptors, Estrogen/metabolism , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Clin Cancer Res ; 20(16): 4314-25, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24947930

ABSTRACT

PURPOSE: Patients with luminal breast cancer (LBC) often become endocrine resistant over time. We investigated the molecular changes associated with acquired hormonoresistances in patient-derived xenografts of LBC. EXPERIMENTAL DESIGN: Two LBC xenografts (HBCx22 and HBCx34) were treated with different endocrine treatments (ET) to obtain xenografts with acquired resistances to tamoxifen (TamR) and ovariectomy (OvaR). PI3K pathway activation was analyzed by Western blot analysis and IHC and responses to ET combined to everolimus were investigated in vivo. Gene expression analyses were performed by RT-PCR and Affymetrix arrays. RESULTS: HBCx22 TamR xenograft was cross-resistant to several hormonotherapies, whereas HBCx22 OvaR and HBCx34 TamR exhibited a treatment-specific resistance profile. PI3K pathway was similarly activated in parental and resistant xenografts but the addition of everolimus did not restore the response to tamoxifen in TamR xenografts. In contrast, the combination of fulvestrant and everolimus induced tumor regression in vivo in HBCx34 TamR, where we found a cross-talk between the estrogen receptor (ER) and PI3K pathways. Expression of several ER-controlled genes and ER coregulators was significantly changed in both TamR and OvaR tumors, indicating impaired ER transcriptional activity. Expression changes associated with hormonoresistance were both tumor and treatment specific and were enriched for genes involved in cell growth, cell death, and cell survival. CONCLUSIONS: PDX models of LBC with acquired resistance to endocrine therapies show a great diversity of resistance phenotype, associated with specific deregulations of ER-mediated gene transcription. These models offer a tool for developing anticancer therapies and to investigate the dynamics of resistance emerging during pharmacologic interventions. Clin Cancer Res; 20(16); 4314-25. ©2014 AACR.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Receptor, ErbB-2/metabolism , Tamoxifen/pharmacology , Animals , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Estrogen Receptor alpha/genetics , Female , Humans , Immunoenzyme Techniques , Mice , Mice, Nude , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, ErbB-2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
PLoS One ; 8(12): e84473, 2013.
Article in English | MEDLINE | ID: mdl-24367664

ABSTRACT

Some strains of West Nile virus (WNV) are neuroinvasive and may induce fatal encephalitis/meningitis in a variety of animal species including humans. Whether, however, there is a strain-specific signature in the brain is as yet unknown. Here we investigated the neuropathogenesis induced by two phylogenetically distant WNV strains of lineage 1, WNV(IS98) and WNV(KUN35 911). While four-week old C57Bl/6J mice were susceptible to both strains and succumbed rapidly after intraperitoneal inoculation, differences were observed in virulence and clinical disease. WNV(KUN35 911), the less virulent strain as judged by determination of LD50, induced typical signs of encephalitis. Such signs were not observed in WNV(IS98)-infected mice, although they died more rapidly. Histological examination of brain sections also revealed differences, as the level of apoptosis and inflammation was higher in WNV(KUN35 911)- than WNV(IS98)-infected mice. Moreover, staining for cleaved caspase 3 showed that the two WNV strains induced apoptotic death through different molecular mechanisms in one particular brain area. Finally, the two strains showed similar tropism in cortex, striatum, brainstem, and cerebellum but a different one in hippocampus. In summary, our data show that, upon peripheral administration, WNV(IS98) and WNV(KUN35 911) strains induce partially distinct lesions and tissue tropism in the brain. They suggest that the virulence of a WNV strain is not necessarily correlated with the severity of apoptotic and inflammatory lesions in the brain.


Subject(s)
Brain/pathology , Brain/virology , West Nile virus/pathogenicity , Animals , Apoptosis , Inflammation/virology , Mice , Mice, Inbred C57BL , Species Specificity , West Nile virus/physiology
13.
Lab Invest ; 93(5): 611-21, 2013 May.
Article in English | MEDLINE | ID: mdl-23459372

ABSTRACT

Metabolic adaptations and changes in the expression of nutrient transporters are known to accompany tumorigenic processes. Nevertheless, in the context of solid tumors, studies of metabolism are hindered by a paucity of tools allowing the identification of cell surface transporters on individual cells. Here, we developed a method for the dissociation of human breast cancer tumor xenografts combined with quantification of cell surface markers, including metabolite transporters. The expression profiles of four relevant nutrient transporters for cancer cells' metabolism, Glut1, ASCT2, PiT1 and PiT2 (participating to glucose, glutamine and inorganic phosphate, respectively), as detected by new retroviral envelope glycoprotein-derived ligands, were distinctive of each tumor, unveiling underlying differences in metabolic pathways. Our tumor dissociation procedure and nutrient transporter profiling technology provides opportunities for future basic research, clinical diagnosis, prognosis and evaluation of therapeutic responses, as well as for drug discovery and development.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Flow Cytometry/methods , Membrane Transport Proteins/metabolism , Analysis of Variance , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Cell Survival/physiology , Female , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Immunohistochemistry/methods , Mice , Mice, Nude , Neoplasm Transplantation , Reproducibility of Results , Transplantation, Heterologous
14.
Mol Cancer Ther ; 11(12): 2693-703, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23012245

ABSTRACT

Triple-negative breast cancers (TNBC) have an aggressive phenotype with a relatively high rate of recurrence and poor overall survival. To date, there is no approved targeted therapy for TNBCs. Aurora kinases act as regulators of mammalian cell division. They are important for cell-cycle progression and are frequently overexpressed or mutated in human tumors, including breast cancer. In this study, we investigated the therapeutic potential of targeting Aurora kinases in preclinical models of human breast cancers using a pan-inhibitor of Aurora kinases, AS703569. In vitro, AS703569 was tested in 15 human breast cancer cell lines. TNBC cell lines were more sensitive to AS703569 than were other types of breast cancer cells. Inhibition of proliferation was associated with cell-cycle arrest, aneuploidy, and apoptosis. In vivo, AS703569 administered alone significantly inhibited tumor growth in seven of 11 patient-derived breast cancer xenografts. Treatment with AS703569 was associated with a decrease of phospho-histone H3 expression. Finally, AS703569 combined to doxorubicin-cyclophosphamide significantly inhibited in vivo tumor recurrence, suggesting that Aurora kinase inhibitors could be used both in monotherapy and in combination settings. In conclusion, these data indicate that targeting Aurora kinases could represent a new effective approach for TNBC treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Aurora Kinases , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Female , Humans , Immunohistochemistry , MCF-7 Cells , Mice , Mice, Nude , Molecular Targeted Therapy , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
15.
BMC Res Notes ; 3: 17, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-20180997

ABSTRACT

BACKGROUND: Expression of several copies of the heat-inducible Hsp70.1Luciferase (LUC) transgene inserted at a single X chromosome locus of a bull (Bos taurus) was assessed in females after X-chromosome inactivation (XCI). Furthermore, impact of the chromosomal environment on the spontaneous expression of these transgene copies before XCI was studied during early development in embryos obtained after in vitro fertilization (IVF), when the locus was carried by the X chromosome inherited from the bull, and after somatic cell nuclear transfer (SCNT) cloning, when the locus could be carried by the inactive Xi or the active Xa chromosome in a female donor cell, or by the (active) X in a male donor cell. FINDINGS: Transgene copies were mapped to bovine Xp22. In XXLUC female fibroblasts, i.e. after random XCI, the proportions of late-replicating inactive and early-replicating active XLUC chromosomes were not biased and the proportion of cells displaying an increase in the level of immunostained luciferase protein after heat-shock induction was similar to that in male fibroblasts. Spontaneous transgene expression occurred at the 8-16-cell stage both in transgenic (female) embryos obtained after IVF and in male and female embryos obtained after SCNT. CONCLUSIONS: The XLUC chromosome is normally inactivated but at least part of the inactivated X-linked Hsp70.1Luciferase transgene copies remains heat-inducible after random XCI in somatic cells. Before XCI, the profile of the transgenes' spontaneous expression is independent of the epigenetic origin of the XLUC chromosome since it is similar in IVF female, SCNT male and SCNT female embryos.

16.
Int. j. morphol ; 24(2): 285-292, jun. 2006. ilus, tab
Article in English | LILACS | ID: lil-432814

ABSTRACT

RESUMEN: Los objetivos de este trabajo fueron los de producir embriones de pudú, obtenidos por la transferencia de núcleos de fibroblastos de la oreja de pudú en ovocitos de un rumiante domésticos que es el bovino. Para posteriormente en un trabajo futuro proceder a la transferencia de embriones de pudú, al útero de hembras receptoras sincronizadas de otra especie. Se obtuvieron biopsias de 1 mm aproximadamente del borde externo de la orejas de dos ciervos pudu machos del jardín zoológico Buin-Zoo, Santiago de Chile. Las líneas celulares han sido establecidas y conservadas según los protocolos utilizados para las bovinos. Los ovocitos son obtenidos por punción del complejo cúmulos-ovocito (COC).desde ovarios de vacas recuperados del matadero. Cada ovocito es enucleado y fusionado con un fibroblasto aislado insertado bajo la zona pelúcida. La fusión de membranas celulares es obtenida por choques eléctricos. En cuanto a la cronología, observamos que al segundo día se forma una etapa de dos blastómeras, al tercer día mórulas de 8 a 16 células, y desde el cuarto día se ha diferenciado como blastocisto, el cuál al séptimo día termina por eclosionar de la zona pelúcida.La obtención de blastocistos embrionarios indica que es posible obtener embriones de pudú mediante clonaje heteroespecífico, aunque, el porcentaje de éxito obtenido es relativamente bajo. Queda aun por verificar la viabilidad de los embriones así obtenidos después de la transferencia in útero.


Subject(s)
Animals , Female , Cattle/embryology , Cattle/genetics , Deer/embryology , Deer/genetics , Cloning, Organism/methods , Cloning, Organism/trends , Insemination, Artificial/methods , Insemination, Artificial , Ruminants/growth & development , Ruminants/embryology
17.
Int. j. morphol ; 23(4): 329-336, 2005. ilus
Article in English | LILACS | ID: lil-626804

ABSTRACT

The somatic cloning by transfer of the nuclei of differentiated adult cells to previously enucleated oocytes is a promising technique for the production of embryos of high genetic value. The better mastering of somatic cloning gives us the possibility to produce embryos from endangered species. The huemul is an Andean native deer, that has been declared an endangered species, it holds a great patrimonial value and it is a Chilean national emblem. In Chile the huemul has the status of protected animal on thirteen Parks and National Reserves managed by Corporacion Nacional Forestal (CONAF). This protection, however, is considered insufficient due to the few geographical connections between the different protected areas. Furthermore, a great proportion of these areas are not subjected to use or they do not constitute adequate habitats. Many authors have proposed that the use of biotechnological methods in reproduction and assisted procreation may help conservational programs orientated to the protection of deer species threatened by extinction. All the anterior prompted us to initiate this study concerning the production of cloned huemul embryos.


El clonaje somático por transferencia del núcleo de células diferenciadas adultas a un ovocito, al que se le ha extraído el núcleo (enucleado), es una técnica prometedora para la producción de embriones de alto valor genético. El mejor dominio del clonaje somático da la posibilidad de producir embriones de especies amenazadas de extinción. El huemul es un ciervo andino autóctono, declarado como especie en peligro de extinción. tiene un gran valor patrimonial, y es emblema de la nación chilena. En este país, el huemul se encuentra protegido en trece Parques y Reservas Nacionales, manejadas por la Corporación Nacional Forestal (CONAF). Sin embargo, su protección se considera insuficiente debido a la baja conectividad entre las áreas protegidas y además, una gran proporción de estas áreas no son utilizadas o no constituyen un hábitat adecuado. Para las especies de cérvidos en vías de extinción el uso de biotecnología reproductiva y métodos de procreación asistida, según varios autores, pueden ayudar a los programas de conservación. Las técnicas clásicas de producción de embriones basados en superovulación, inseminación artificial y transferencia embrionaria,en los cérvidos, han resultado muy dificultosa. Esto, sumado a las características del huemul, que no permite su estabulación en cautiverio, nos ha movido a iniciar un estudio tendiente a la producción de embriones clonados de esta especie.


Subject(s)
Animals , Deer/genetics , Endangered Species , Cloning, Organism , Chile
SELECTION OF CITATIONS
SEARCH DETAIL
...