Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 14 de 14
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Nat Med ; 30(4): 990-1000, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38605166

RÉSUMÉ

Children with rare, relapsed or refractory cancers often face limited treatment options, and few predictive biomarkers are available that can enable personalized treatment recommendations. The implementation of functional precision medicine (FPM), which combines genomic profiling with drug sensitivity testing (DST) of patient-derived tumor cells, has potential to identify treatment options when standard-of-care is exhausted. The goal of this prospective observational study was to generate FPM data for pediatric patients with relapsed or refractory cancer. The primary objective was to determine the feasibility of returning FPM-based treatment recommendations in real time to the FPM tumor board (FPMTB) within a clinically actionable timeframe (<4 weeks). The secondary objective was to assess clinical outcomes from patients enrolled in the study. Twenty-five patients with relapsed or refractory solid and hematological cancers were enrolled; 21 patients underwent DST and 20 also completed genomic profiling. Median turnaround times for DST and genomics were within 10 days and 27 days, respectively. Treatment recommendations were made for 19 patients (76%), of whom 14 received therapeutic interventions. Six patients received subsequent FPM-guided treatments. Among these patients, five (83%) experienced a greater than 1.3-fold improvement in progression-free survival associated with their FPM-guided therapy relative to their previous therapy, and demonstrated a significant increase in progression-free survival and objective response rate compared to those of eight non-guided patients. The findings from our proof-of-principle study illustrate the potential for FPM to positively impact clinical care for pediatric and adolescent patients with relapsed or refractory cancers and warrant further validation in large prospective studies. ClinicalTrials.gov registration: NCT03860376 .


Sujet(s)
Tumeurs hématologiques , Tumeurs , Adolescent , Enfant , Humains , Médecine de précision , Études prospectives , Études de faisabilité , Tumeurs/génétique , Tumeurs/thérapie
2.
Pharmacol Ther ; 234: 108048, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-34848203

RÉSUMÉ

Translocator Protein 18 kDa (TSPO), previously named Peripheral Benzodiazepine Receptor, is a well-validated and widely used biomarker of neuroinflammation to assess diverse central nervous system (CNS) pathologies in preclinical and clinical studies. Many studies have shown that in animal models of human neurological and neurodegenerative disease and in the human condition, TSPO levels increase in the brain neuropil, and this increase is driven by infiltration of peripheral inflammatory cells and activation of glial cells. Therefore, a clear understanding of the dynamics of the cellular sources of the TSPO response is critically important in the interpretation of Positron Emission Tomography (PET) studies and for understanding the pathophysiology of CNS diseases. Within the normal brain compartment, there are tissues and cells such as the choroid plexus, ependymal cells of the lining of the ventricles, and vascular endothelial cells that also express TSPO at even higher levels than in glial cells. However, there is a paucity of knowledge if these cell types respond and increase TSPO in the diseased brain. These cells do provide a background signal that needs to be accounted for in TSPO-PET imaging studies. More recently, there are reports that TSPO may be expressed in neurons of the adult brain and TSPO expression may be increased by neuronal activity. Therefore, it is essential to study this topic with a great deal of detail, methodological rigor, and rule out alternative interpretations and imaging artifacts. High levels of TSPO are present in the outer mitochondrial membrane. Recent studies have provided evidence of its localization in other cellular compartments including the plasma membrane and perinuclear regions which may define functions that are different from that in mitochondria. A greater understanding of the TSPO subcellular localization in glial cells and infiltrating peripheral immune cells and associated function(s) may provide an additional layer of information to the understanding of TSPO neurobiology. This review is an effort to outline recent advances in understanding the cellular sources and subcellular localization of TSPO in brain cells and to examine remaining questions that require rigorous investigation.


Sujet(s)
Maladies neurodégénératives , Récepteurs GABA , Animaux , Encéphale/imagerie diagnostique , Encéphale/métabolisme , Cellules endothéliales/métabolisme , Humains , Maladies neurodégénératives/métabolisme , Maladies neuro-inflammatoires , Tomographie par émission de positons/méthodes , Récepteurs GABA/métabolisme
4.
Cancers (Basel) ; 13(18)2021 Sep 08.
Article de Anglais | MEDLINE | ID: mdl-34572751

RÉSUMÉ

Glioblastoma (GBM) is the most common primary brain tumor in adults, with few available therapies and a five-year survival rate of 7.2%. Hence, strategies for improving GBM prognosis are urgently needed. The translocator protein 18kDa (TSPO) plays crucial roles in essential mitochondria-based physiological processes and is a validated biomarker of neuroinflammation, which is implicated in GBM progression. The TSPO gene has a germline single nucleotide polymorphism, rs6971, which is the most common SNP in the Caucasian population. High TSPO gene expression is associated with reduced survival in GBM patients; however, the relation between the most frequent TSPO genetic variant and GBM pathogenesis is not known. The present study retrospectively analyzed the correlation of the TSPO polymorphic variant rs6971 with overall and progression-free survival in GBM patients using three independent cohorts. TSPO rs6971 polymorphism was significantly associated with shorter overall survival and progression-free survival in male GBM patients but not in females in one large cohort of 441 patients. We observed similar trends in two other independent cohorts. These observations suggest that the TSPO rs6971 polymorphism could be a significant predictor of poor prognosis in GBM, with a potential for use as a prognosis biomarker in GBM patients. These results reveal for the first time a biological sex-specific relation between rs6971 TSPO polymorphism and GBM.

5.
J Leukoc Biol ; 110(1): 123-140, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-33205494

RÉSUMÉ

Translocator protein 18 kDa (TSPO) is a well-known outer mitochondrial membrane protein and it is widely used as a biomarker of neuroinflammation and brain injury. Although it is thought that TSPO plays key roles in a multitude of host cell functions, including steroid biosynthesis, apoptosis, generation of reactive oxygen species, and proliferation, some of these functions have recently been questioned. Here, we report the unexpected finding that circulating immune cells differentially express basal levels of TSPO on their cell surface, with a high percentage of monocytes and neutrophils expressing cell surface TSPO. In vitro stimulation of monocytes with LPS significantly increases the frequency of cells with surface TSPO expression in the absence of altered gene expression. Importantly, the LPS increase in TSPO cell surface expression in monocytes appears to be selective for LPS because two other distinct monocyte activators failed to increase the frequency of cells with surface TSPO. Finally, when we quantified immune cell TSPO surface expression in antiretroviral therapy-treated HIV+ donors, a chronic inflammatory disease, we found significant increases in the frequency of TSPO surface localization, which could be pharmacologically suppressed with ∆9 -tetrahydrocannabinol. These findings suggest that cell surface TSPO in circulating leukocytes could serve as a peripheral blood-based biomarker of inflammation.


Sujet(s)
Infections à VIH/immunologie , Infections à VIH/métabolisme , Infections à VIH/virologie , Lipopolysaccharides/effets indésirables , Récepteurs GABA/métabolisme , Animaux , Thérapie antirétrovirale hautement active , Apoptose , Marqueurs biologiques , Prédisposition aux maladies , Infections à VIH/traitement médicamenteux , Humains , Inflammation/étiologie , Inflammation/métabolisme , Leucocytes/immunologie , Leucocytes/métabolisme , Mitochondries/génétique , Mitochondries/métabolisme , Transport des protéines , Espèces réactives de l'oxygène/métabolisme , Récepteurs GABA/génétique
6.
Mol Neurobiol ; 57(11): 4467-4487, 2020 Nov.
Article de Anglais | MEDLINE | ID: mdl-32743737

RÉSUMÉ

In the brain neuropil, translocator protein 18 kDa (TSPO) is a stress response protein that is upregulated in microglia and astrocytes in diverse central nervous system pathologies. TSPO is widely used as a biomarker of neuroinflammation in preclinical and clinical neuroimaging studies. However, there is a paucity of knowledge on the function(s) of TSPO in glial cells. In this study, we explored a putative interaction between TSPO and NADPH oxidase 2 (NOX2) in microglia. We found that TSPO associates with gp91phox and p22phox, the principal subunits of NOX2 in primary murine microglia. The association of TSPO with gp91phox and p22phox was observed using co-immunoprecipitation, confocal immunofluorescence imaging, and proximity ligation assay. We found that besides gp91phox and p22phox, voltage-dependent anion channel (VDAC) also co-immunoprecipitated with TSPO consistent with previous reports. When we compared lipopolysaccharide (LPS) stimulated microglia to vehicle control, we found that a lower amount of gp91phox and p22phox protein co-immunoprecipitated with TSPO suggesting a disruption of the TSPO-NOX2 subunits association. TSPO immuno-gold electron microscopy confirmed that TSPO is present in the outer mitochondrial membrane but it is also found in the endoplasmic reticulum (ER), mitochondria-associated ER membrane (MAM), and in the plasma membrane. TSPO localization at the MAM may represent a subcellular site where TSPO interacts with gp91phox and p22phox since the MAM is a point of communication between outer mitochondria membrane proteins (TSPO) and ER proteins (gp91phox and p22phox) where they mature and form the cytochrome b558 (Cytb558) heterodimer. We also found that an acute burst of reactive oxygen species (ROS) increased TSPO levels on the surface of microglia and this effect was abrogated by a ROS scavenger. These results suggest that ROS production may alter the subcellular distribution of TSPO. Collectively, our findings suggest that in microglia, TSPO is associated with the major NOX2 subunits gp91phox and p22phox. We hypothesize that this interaction may regulate Cytb558 formation and modulate NOX2 levels, ROS production, and redox homeostasis in microglia.


Sujet(s)
Microglie/métabolisme , NADPH oxidase/métabolisme , Récepteurs GABA/métabolisme , Animaux , Cytosol/métabolisme , Réticulum endoplasmique/métabolisme , Femelle , Cellules HEK293 , Hème/métabolisme , Humains , Membranes intracellulaires/métabolisme , Souris de lignée C57BL , Souris transgéniques , Microglie/ultrastructure , Mitochondries/métabolisme , Modèles biologiques , Porphyrines/métabolisme , Liaison aux protéines , Espèces réactives de l'oxygène/métabolisme , Récepteurs GABA/composition chimique , Canaux anioniques voltage-dépendants/métabolisme
7.
Anticancer Res ; 39(8): 4023-4030, 2019 Aug.
Article de Anglais | MEDLINE | ID: mdl-31366484

RÉSUMÉ

BACKGROUND: Treatment options for patients with platinum-resistant ovarian cancer are generally palliative in nature and rarely have realistic potential to be curative. Because many patients with recurrent ovarian cancer receive aggressive chemotherapy for prolonged periods, sometimes continuously, therapy-related toxicities are a major factor in treatment decisions. The use of ex vivo drug sensitivity screens has the potential to improve the treatment of patients with platinum-resistant ovarian cancer by providing personalized treatment plans and thus reducing toxicity from unproductive therapy attempts. MATERIALS AND METHODS: We evaluated the treatment responses of a set of six early-passage patient-derived ovarian cancer cell lines towards a set of 30 Food and Drug Administration-approved chemotherapy drugs using drug-sensitivity testing. RESULTS: We observed a wide range of treatment responses of the cell lines. While most compounds displayed vastly different treatment responses between cell lines, we found that some compounds such as docetaxel and cephalomannine reduced cell survival of all cell lines. CONCLUSION: We propose that ex vivo drug-sensitivity screening holds the potential to greatly improve patient outcomes, especially in a population where multiple continuous treatments are not an option due to advanced disease, rapid disease progression, age or poor overall health. This approach may also be useful to identify potential novel therapeutics for patients with ovarian cancer.


Sujet(s)
Résistance aux médicaments antinéoplasiques/génétique , Récidive tumorale locale/traitement médicamenteux , Tumeurs de l'ovaire/traitement médicamenteux , Platine/pharmacologie , Antinéoplasiques/effets indésirables , Antinéoplasiques/pharmacologie , Protocoles de polychimiothérapie antinéoplasique , Survie cellulaire/effets des médicaments et des substances chimiques , Femelle , Humains , Récidive tumorale locale/anatomopathologie , Tumeurs de l'ovaire/anatomopathologie , Ovaire/effets des médicaments et des substances chimiques , Ovaire/anatomopathologie , Platine/effets indésirables
8.
Clin Cancer Res ; 24(19): 4874-4886, 2018 10 01.
Article de Anglais | MEDLINE | ID: mdl-29959144

RÉSUMÉ

Purpose: Rational targeted therapies are needed for treatment of ovarian cancers. Signaling kinases Src and MAPK are activated in high-grade serous ovarian cancer (HGSOC). Here, we tested the frequency of activation of both kinases in HGSOC and the therapeutic potential of dual kinase inhibition.Experimental Design: MEK and Src activation was assayed in primary HGSOC from The Cancer Genome Atlas (TGGA). Effects of dual kinase inhibition were assayed on cell-cycle, apoptosis, gene, and proteomic analysis; cancer stem cells; and xenografts.Results: Both Src and MAPK are coactivated in 31% of HGSOC, and this associates with worse overall survival on multivariate analysis. Frequent dual kinase activation in HGSOC led us to assay the efficacy of combined Src and MEK inhibition. Treatment of established lines and primary ovarian cancer cultures with Src and MEK inhibitors saracatinib and selumetinib, respectively, showed target kinase inhibition and synergistic induction of apoptosis and cell-cycle arrest in vitro, and tumor inhibition in xenografts. Gene expression and proteomic analysis confirmed cell-cycle inhibition and autophagy. Dual therapy also potently inhibited tumor-initiating cells. Src and MAPK were both activated in tumor-initiating populations. Combination treatment followed by drug washout decreased sphere formation and ALDH1+ cells. In vivo, tumors dissociated after dual therapy showed a marked decrease in ALDH1 staining, sphere formation, and loss of tumor-initiating cells upon serial xenografting.Conclusions: Selumetinib added to saracatinib overcomes EGFR/HER2/ERBB2-mediated bypass activation of MEK/MAPK observed with saracatinib alone and targets tumor-initiating ovarian cancer populations, supporting further evaluation of combined Src-MEK inhibition in clinical trials. Clin Cancer Res; 24(19); 4874-86. ©2018 AACR.


Sujet(s)
MAP Kinase Kinase 1/antagonistes et inhibiteurs , Tumeurs de l'ovaire/traitement médicamenteux , Protéomique , src-Family kinases/antagonistes et inhibiteurs , Animaux , Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Apoptose/effets des médicaments et des substances chimiques , Benzimidazoles/pharmacologie , Benzodioxoles/administration et posologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie sans rechute , Résistance aux médicaments antinéoplasiques , Récepteur alpha des oestrogènes/génétique , Femelle , Humains , MAP Kinase Kinase 1/génétique , Souris , Adulte d'âge moyen , Cellules souches tumorales/effets des médicaments et des substances chimiques , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Inhibiteurs de protéines kinases/administration et posologie , Quinazolines/administration et posologie , Tests d'activité antitumorale sur modèle de xénogreffe , src-Family kinases/génétique
9.
Mol Cancer Res ; 15(2): 152-164, 2017 02.
Article de Anglais | MEDLINE | ID: mdl-28108626

RÉSUMÉ

Resistance to hormonal therapies is a major clinical problem in the treatment of estrogen receptor α-positive (ERα+) breast cancers. Epigenetic marks, namely DNA methylation of cytosine at specific CpG sites (5mCpG), are frequently associated with ERα+ status in human breast cancers. Therefore, ERα may regulate gene expression in part via DNA methylation. This hypothesis was evaluated using a panel of breast cancer cell line models of antiestrogen resistance. Microarray gene expression profiling was used to identify genes normally silenced in ERα+ cells but derepressed upon exposure to the demethylating agent decitabine, derepressed upon long-term loss of ERα expression, and resuppressed by gain of ERα activity/expression. ERα-dependent DNA methylation targets (n = 39) were enriched for ERα-binding sites, basal-up/luminal-down markers, cancer stem cell, epithelial-mesenchymal transition, and inflammatory and tumor suppressor genes. Kaplan-Meier survival curve and Cox proportional hazards regression analyses indicated that these targets predicted poor distant metastasis-free survival among a large cohort of breast cancer patients. The basal breast cancer subtype markers LCN2 and IFI27 showed the greatest inverse relationship with ERα expression/activity and contain ERα-binding sites. Thus, genes that are methylated in an ERα-dependent manner may serve as predictive biomarkers in breast cancer. IMPLICATIONS: ERα directs DNA methylation-mediated silencing of specific genes that have biomarker potential in breast cancer subtypes. Mol Cancer Res; 15(2); 152-64. ©2016 AACR.


Sujet(s)
Tumeurs du sein/génétique , Méthylation de l'ADN , Transition épithélio-mésenchymateuse/génétique , Récepteur alpha des oestrogènes/génétique , Cellules souches tumorales/physiologie , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Récepteur alpha des oestrogènes/biosynthèse , Femelle , Régulation de l'expression des gènes tumoraux , Extinction de l'expression des gènes , Humains , Cellules MCF-7
10.
Cancer Res ; 76(2): 491-504, 2016 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-26744520

RÉSUMÉ

Consequences of the obesity epidemic on cancer morbidity and mortality are not fully appreciated. Obesity is a risk factor for many cancers, but the mechanisms by which it contributes to cancer development and patient outcome have yet to be fully elucidated. Here, we examined the effects of coculturing human-derived adipocytes with established and primary breast cancer cells on tumorigenic potential. We found that the interaction between adipocytes and cancer cells increased the secretion of proinflammatory cytokines. Prolonged culture of cancer cells with adipocytes or cytokines increased the proportion of mammosphere-forming cells and of cells expressing stem-like markers in vitro. Furthermore, contact with immature adipocytes increased the abundance of cancer cells with tumor-forming and metastatic potential in vivo. Mechanistic investigations demonstrated that cancer cells cultured with immature adipocytes or cytokines activated Src, thus promoting Sox2, c-Myc, and Nanog upregulation. Moreover, Sox2-dependent induction of miR-302b further stimulated cMYC and SOX2 expression and potentiated the cytokine-induced cancer stem cell-like properties. Finally, we found that Src inhibitors decreased cytokine production after coculture, indicating that Src is not only activated by adipocyte or cytokine exposures, but is also required to sustain cytokine induction. These data support a model in which cancer cell invasion into local fat would establish feed-forward loops to activate Src, maintain proinflammatory cytokine production, and increase tumor-initiating cell abundance and metastatic progression. Collectively, our findings reveal new insights underlying increased breast cancer mortality in obese individuals and provide a novel preclinical rationale to test the efficacy of Src inhibitors for breast cancer treatment.


Sujet(s)
Adipocytes/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Cytokines/métabolisme , Obésité/complications , ARN messager/métabolisme , src-Family kinases/métabolisme , Adipocytes/cytologie , Animaux , Tumeurs du sein/anatomopathologie , Évolution de la maladie , Femelle , Humains , Souris , ARN messager/génétique , Facteurs de transcription SOX-B1 , Transduction du signal , Transfection , src-Family kinases/génétique
11.
Cell ; 159(3): 499-513, 2014 Oct 23.
Article de Anglais | MEDLINE | ID: mdl-25417103

RÉSUMÉ

Stromal communication with cancer cells can influence treatment response. We show that stromal and breast cancer (BrCa) cells utilize paracrine and juxtacrine signaling to drive chemotherapy and radiation resistance. Upon heterotypic interaction, exosomes are transferred from stromal to BrCa cells. RNA within exosomes, which are largely noncoding transcripts and transposable elements, stimulates the pattern recognition receptor RIG-I to activate STAT1-dependent antiviral signaling. In parallel, stromal cells also activate NOTCH3 on BrCa cells. The paracrine antiviral and juxtacrine NOTCH3 pathways converge as STAT1 facilitates transcriptional responses to NOTCH3 and expands therapy-resistant tumor-initiating cells. Primary human and/or mouse BrCa analysis support the role of antiviral/NOTCH3 pathways in NOTCH signaling and stroma-mediated resistance, which is abrogated by combination therapy with gamma secretase inhibitors. Thus, stromal cells orchestrate an intricate crosstalk with BrCa cells by utilizing exosomes to instigate antiviral signaling. This expands BrCa subpopulations adept at resisting therapy and reinitiating tumor growth.


Sujet(s)
Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/radiothérapie , Exosomes/métabolisme , Communication paracrine , Cellules stromales/métabolisme , Animaux , Tumeurs du sein/métabolisme , Lignée cellulaire tumorale , Simulation numérique , Résistance aux médicaments antinéoplasiques , Femelle , Humains , Interférons/métabolisme , Souris nude , Radiotolérance , Récepteurs Notch/métabolisme , Facteur de transcription STAT-1/métabolisme , Transduction du signal , Protéines G rab/métabolisme
12.
Breast Cancer Res Treat ; 144(3): 503-17, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24567196

RÉSUMÉ

Our goal was to establish primary cultures from dissociation of breast tumors in order to provide cellular models that may better recapitulate breast cancer pathogenesis and the metastatic process. Here, we report the characterization of six cellular models derived from the dissociation of primary breast tumor specimens, referred to as "dissociated tumor (DT) cells." In vitro, DT cells were characterized by proliferation assays, colony formation assays, protein, and gene expression profiling, including PAM50 predictor analysis. In vivo, tumorigenic and metastatic potential of DT cultures was assessed in NOD/SCID and NSG mice. These cellular models differ from recently developed patient-derived xenograft models in that they can be used for both in vitro and in vivo studies. PAM50 predictor analysis showed DT cultures similar to their paired primary tumor and as belonging to the basal and Her2-enriched subtypes. In vivo, three DT cultures are tumorigenic in NOD/SCID and NSG mice, and one of these is metastatic to lymph nodes and lung after orthotopic inoculation into the mammary fat pad, without excision of the primary tumor. Three DT cultures comprised of cancer-associated fibroblasts (CAFs) were isolated from luminal A, Her2-enriched, and basal primary tumors. Among the DT cells are those that are tumorigenic and metastatic in immunosuppressed mice, offering novel cellular models of ER-negative breast cancer subtypes. A group of CAFs provide tumor subtype-specific components of the tumor microenvironment (TME). Altogether, these DT cultures provide closer-to-primary cellular models for the study of breast cancer pathogenesis, metastasis, and TME.


Sujet(s)
Tumeurs du sein/anatomopathologie , Culture de cellules primaires , Animaux , Marqueurs biologiques tumoraux/métabolisme , Tumeurs du sein/diagnostic , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Cycle cellulaire , Prolifération cellulaire , Transformation cellulaire néoplasique , Modèles animaux de maladie humaine , Femelle , Fibroblastes/anatomopathologie , Analyse de profil d'expression de gènes , Hétérogreffes , Humains , Immunohistochimie , Souris , Métastase tumorale , Culture de cellules primaires/méthodes , Charge tumorale , Cellules cancéreuses en culture , Test clonogénique de cellules souches tumorales
13.
EMBO Mol Med ; 5(10): 1502-22, 2013 10.
Article de Anglais | MEDLINE | ID: mdl-23982961

RÉSUMÉ

Increasing evidence suggests that stem-like cells mediate cancer therapy resistance and metastasis. Breast tumour-initiating stem cells (T-ISC) are known to be enriched in CD44(+) CD24(neg/low) cells. Here, we identify two T-ISC subsets within this population in triple negative breast cancer (TNBC) lines and dissociated primary breast cancer cultures: CD44(+) CD24(low+) subpopulation generates CD44(+) CD24(neg) progeny with reduced sphere formation and tumourigenicity. CD44(+) CD24(low+) populations contain subsets of ALDH1(+) and ESA(+) cells, yield more frequent spheres and/or T-ISC in limiting dilution assays, preferentially express metastatic gene signatures and show greater motility, invasion and, in the MDA-MB-231 model, metastatic potential. CD44(+) CD24(low+) but not CD44(+) CD24(neg) express activated Notch1 intracellular domain (N1-ICD) and Notch target genes. We show N1-ICD transactivates SOX2 to increase sphere formation, ALDH1+ and CD44(+) CD24(low+) cells. Gamma secretase inhibitors (GSI) reduced sphere formation and xenograft growth from CD44(+) CD24(low+) cells, but CD44(+) CD24(neg) were resistant. While GSI hold promise for targeting T-ISC, stem cell heterogeneity as observed herein, could limit GSI efficacy. These data suggest a breast T-ISC hierarchy in which distinct pathways drive developmentally related subpopulations with different anti-cancer drug responsiveness.


Sujet(s)
Amyloid precursor protein secretases/antagonistes et inhibiteurs , Aldéhyde déshydrogénase-1 , Amyloid precursor protein secretases/métabolisme , Animaux , Antinéoplasiques/composition chimique , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/toxicité , Antigènes CD24/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Antienzymes/composition chimique , Antienzymes/usage thérapeutique , Antienzymes/toxicité , Femelle , Humains , Antigènes CD44/métabolisme , Isoenzymes/métabolisme , Cellules MCF-7 , Souris , Souris de lignée BALB C , Souris nude , Cellules souches tumorales/cytologie , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Récepteurs Notch/métabolisme , Retinal dehydrogenase/métabolisme , Facteurs de transcription SOX-B1/antagonistes et inhibiteurs , Facteurs de transcription SOX-B1/génétique , Facteurs de transcription SOX-B1/métabolisme , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Cellules cancéreuses en culture
14.
Article de Anglais | MEDLINE | ID: mdl-17901003

RÉSUMÉ

A rapid, reproducible and accurate high-performance liquid chromatographic (HPLC) method for the quantitative determination of sphingomyelin in rat brain was developed and validated using normal-phase silica gel column, acetonitrile-methanol-water (65:18:17 (v/v)) at a flow rate of 1 ml/min, isocratic elution, UV detection at 207 nm and 1,2-dimyristoyl-sn-glycero-3-phosphocholine as an internal standard. Total run time was 10.0 min. The calibration curve was linear over the range of 0.025-0.4 mg/ml sphingomyelin (R2>0.99). The intra-day coefficient of variation ranged from 1.4% to 2.2%. The average inter-day coefficient of variation over a period of 4 days was 3.1%. The practical limit of detection was 0.005 mg/ml with a quantification limit of 0.01 mg/ml.


Sujet(s)
Chimie du cerveau , Chromatographie en phase liquide à haute performance/méthodes , Sphingomyéline/analyse , Animaux , Dimyristoylphosphatidylcholine/normes , Rats , Normes de référence , Sensibilité et spécificité
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE