Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
1.
Toxicol Sci ; 188(2): 208-218, 2022 07 28.
Article de Anglais | MEDLINE | ID: mdl-35639956

RÉSUMÉ

For all the promise of and need for clinical drug-induced liver injury (DILI) risk screening systems, demonstrating the predictive value of these systems versus readily available physicochemical properties and inherent dosing information has not been thoroughly evaluated. Therefore, we utilized a systematic approach to evaluate the predictive value of in vitro safety assays including bile salt export pump transporter inhibition and cytotoxicity in HepG2 and transformed human liver epithelial along with physicochemical properties. We also evaluated the predictive value of in vitro ADME assays including hepatic partition coefficient (Kp) and its unbound counterpart because they provide insight on hepatic accumulation potential. The datasets comprised of 569 marketed drugs with FDA DILIrank annotation (most vs less/none), dose and physicochemical information, 384 drugs with Kp and plasma protein binding data, and 279 drugs with safety assay data. For each dataset and combination of input parameters, we developed random forest machine learning models and measured model performance using the receiver operator characteristic area under the curve (ROC AUC). The median ROC AUC across the various data and parameters sets ranged from 0.67 to 0.77 with little evidence of additive predictivity when including safety or ADME assay data. Subsequent machine learning models consistently demonstrated daily dose, fraction sp3 or ionization, and cLogP/D inputs produced the best, simplest model for predicting clinical DILI risk with an ROC AUC of 0.75. This systematic framework should be used for future assay predictive value assessments and highlights the need for continued improvements to clinical DILI risk annotation.


Sujet(s)
Lésions hépatiques dues aux substances , Aire sous la courbe , Dosage biologique , Lésions hépatiques dues aux substances/diagnostic , Lésions hépatiques dues aux substances/étiologie , Humains
2.
Toxicol In Vitro ; 70: 105012, 2021 Feb.
Article de Anglais | MEDLINE | ID: mdl-33049313

RÉSUMÉ

Alveolar type II (ATII) epithelial cells contain lamellar bodies (LBs) which synthesize and store lung surfactants. In animals, the inhibition or knockout of leucine-rich repeat kinase 2 (LRRK2) causes abnormal enlargement of LBs in ATII cells. This effect of LRRK2 inhibition in lung is largely accepted as being mediated directly through blocking of the kinase function; however, downstream consequences in the lung remain unknown. In this work we established an in vitro alveolar epithelial cell (AEC) model that recapitulates the in vivo phenotype of ATII cells and developed an assay to quantify changes in LB size in response to LRRK2 inhibitors. Culture of primary human AECs at the air-liquid interface on matrigel and collagen-coated transwell inserts in the presence of growth factors promoted the LB formation and apical microvilli and induced expression of LRRK2 and ATII cell markers. Treatment with a selective LRRK2 inhibitor resulted in pharmacological reduction of phospho-LRRK2 and a significant increase in LB size; effects previously reported in lungs of non-human primates treated with LRRK2 inhibitor. In summary, our human in vitro AEC model recapitulates the abnormal lung findings observed in LRRK2-perturbed animals and holds the potential for expanding current understanding of LRRK2 function in the lung.


Sujet(s)
Pneumocytes/anatomopathologie , Leucine-rich repeat serine-threonine protein kinase-2/antagonistes et inhibiteurs , Modèles biologiques , Transporteurs ABC/métabolisme , Adénocarcinome pulmonaire/métabolisme , Pneumocytes/métabolisme , Pneumocytes/ultrastructure , Cellules cultivées , Évaluation préclinique de médicament , Expression des gènes , Humains , Leucine-rich repeat serine-threonine protein kinase-2/génétique , Leucine-rich repeat serine-threonine protein kinase-2/métabolisme , Tumeurs du poumon/métabolisme , Protéine C associée au surfactant pulmonaire/métabolisme
3.
Arch Toxicol ; 95(1): 149-168, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-32816093

RÉSUMÉ

Preventing clinical drug-induced liver injury (DILI) remains a major challenge, because DILI develops via multifactorial mechanisms. Immune and inflammatory reactions are considered important mechanisms of DILI; however, biomarkers from in vitro systems using immune cells have not been comprehensively studied. The aims of this study were (1) to identify promising biomarker genes for predicting DILI in an in vitro coculture model of peripheral blood mononuclear cells (PBMCs) with a human liver cell line, and (2) to evaluate these genes as predictors of DILI using a panel of drugs with different clinical DILI risk. Transcriptome-wide analysis of PBMCs cocultured with HepG2 or differentiated HepaRG cells that were treated with several drugs revealed an appropriate separation of DILI-positive and DILI-negative drugs, from which 12 putative biomarker genes were selected. To evaluate the predictive performance of these genes, PBMCs cocultured with HepG2 cells were exposed to 77 different drugs, and gene expression levels in PBMCs were determined. The MET proto-oncogene receptor tyrosine kinase (MET) showed the highest area under the receiver-operating characteristic curve (AUC) value of 0.81 among the 12 genes with a high sensitivity/specificity (85/66%). However, a stepwise logistic regression model using the 12 identified genes showed the highest AUC value of 0.94 with a high sensitivity/specificity (93/86%). Taken together, we established a coculture system using PBMCs and HepG2 cells and selected biomarkers that can predict DILI risk. The established model would be useful in detecting the DILI potential of compounds, in particular those that involve an immune mechanism.


Sujet(s)
Lésions hépatiques dues aux substances/étiologie , Hépatocytes/effets des médicaments et des substances chimiques , Agranulocytes/effets des médicaments et des substances chimiques , Transcriptome/effets des médicaments et des substances chimiques , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Survie cellulaire/effets des médicaments et des substances chimiques , Lésions hépatiques dues aux substances/génétique , Lésions hépatiques dues aux substances/immunologie , Lésions hépatiques dues aux substances/métabolisme , Techniques de coculture , Analyse de profil d'expression de gènes , Marqueurs génétiques , Cellules HepG2 , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Humains , Agranulocytes/immunologie , Agranulocytes/métabolisme , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Séquençage par oligonucléotides en batterie , Proto-oncogène Mas , Appréciation des risques
4.
Toxicol Sci ; 161(1): 58-75, 2018 01 01.
Article de Anglais | MEDLINE | ID: mdl-28973697

RÉSUMÉ

Pharmaceuticals and chemicals produce hemangiosarcomas (HS) in mice, often by nongenotoxic, proliferative mechanisms. A mode-of-action (MOA) for hemangiosarcoma was proposed based on information presented at an international workshop (Cohen et al., Hemangiosarcoma in rodents: Mode-of-action evaluation and human relevance. Toxicol. Sci. 111, 4-18.). Five key elements of the MOA were articulated and included hypoxia, macrophage activation, increased angiogenic growth factors, dysregulated angiogenesis/erythropoiesis, and endothial cell proliferation. The goal of the current study was to add to the weight-of-evidence for the proposed MOA by assessing these key elements with 3 different compounds of varying potency for HS induction: fenretinide (high), troglitazone (intermediate), and elmiron (low). Multiple endpoints, including hypoxia (hyproxyprobe, transcriptomics), endothelial cell (EC) proliferation, and clinical and anatomic pathology, were assessed after 2, 4, and 13-weeks of treatment in B6C3F1 mice. All 3 compounds demonstrated strong evidence for dysregulated erythropoiesis (decrease in RBC and a failure to increase reticulocytes) and macrophage activation (4- to 11-fold increases); this pattern of hematological changes in mice might serve as an early biomarker to evaluate EC proliferation in suspected target organs for potential HS formation. Fenretinide demonstrated all 5 key elements, while troglitazone demonstrated 4 and elmiron demonstrated 3. Transcriptomics provided support for the 5 elements of the MOA, but was not any more sensitive than hypoxyprobe immunohistochemistry for detecting hypoxia. The overall transcriptional evidence for the key elements of the proposed MOA was also consistent with the potency of HS induction. These data, coupled with the previous work with 2-butoxyethanol and pregablin, increase the weight-of-evidence for the proposed MOA for HS formation.


Sujet(s)
Fenrétinide/toxicité , Hémangiosarcome/induit chimiquement , Néovascularisation pathologique/induit chimiquement , Pentosane polysulfate/toxicité , Troglitazone/toxicité , Animaux , Hypoxie cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Cellules endothéliales/effets des médicaments et des substances chimiques , Hémangiosarcome/métabolisme , Hémangiosarcome/anatomopathologie , Activation des macrophages/effets des médicaments et des substances chimiques , Mâle , Souris , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/anatomopathologie , Spécificité d'organe
5.
Regul Toxicol Pharmacol ; 91: 93-102, 2017 Dec.
Article de Anglais | MEDLINE | ID: mdl-29074274

RÉSUMÉ

Tofacitinib is an oral JAK inhibitor for the treatment of rheumatoid arthritis. In the 2-year carcinogenicity study with tofacitinib, increased incidence of hibernoma (a neoplasm of brown adipose tissue [BAT]) was noted in female rats at ≥30 mg/kg/day (≥41x human exposure multiples). Thus, signaling pathways within BAT were investigated by measuring BAT: weight, cell proliferation biomarkers, content of basal and prolactin-induced phosphorylated Signal Transducer and Activator of Transcription (STAT), and uncoupling protein 1 (UCP-1). The relationship between cardiovascular hemodynamics and plasma norepinephrine (NE) levels was also investigated. Tofacitinib administered to female rats at doses of 10, 30, or 75 mg/kg/day for 14 days increased BAT weight at 75 mg/kg/day and cell proliferation at ≥30 mg/kg/day. JAK inhibition, observed as lower pSTAT3 and pSTAT5 in BAT, was noted at ≥10 mg/kg/day, while lower activity of BAT was observed as lower UCP-1 protein at ≥30 mg/kg/day. In cultured brown adipocytes, prolactin-induced increase in pSTAT5 and pSTAT3 were inhibited by tofacitinib in a concentration-dependent manner. Tofacitinib lowered blood pressure, increased heart rate, and resulted in dose-dependent increases in circulating NE. Thus, JAK/STAT inhibition in BAT and sympathetic stimulation are two factors which might contribute to the genesis of hibernomas by tofacitinib in rats.


Sujet(s)
Lipome/induit chimiquement , Pipéridines/effets indésirables , Inhibiteurs de protéines kinases/effets indésirables , Pyrimidines/effets indésirables , Pyrroles/effets indésirables , Tissu adipeux brun/effets des médicaments et des substances chimiques , Tissu adipeux brun/métabolisme , Animaux , Pression sanguine/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Rythme cardiaque/effets des médicaments et des substances chimiques , Inhibiteurs des Janus kinases/effets indésirables , Inhibiteurs des Janus kinases/pharmacologie , Lipome/métabolisme , Mâle , Pipéridines/pharmacologie , Inhibiteurs de protéines kinases/pharmacologie , Pyrimidines/pharmacologie , Pyrroles/pharmacologie , Rats , Rat Sprague-Dawley , Facteurs de transcription STAT/antagonistes et inhibiteurs , Transduction du signal/effets des médicaments et des substances chimiques , Transactivateurs/effets indésirables , Transactivateurs/pharmacologie
6.
Toxicol Sci ; 159(1): 42-49, 2017 09 01.
Article de Anglais | MEDLINE | ID: mdl-28903491

RÉSUMÉ

Drug-induced vascular injury (DIVI) in preclinical studies can delay, if not terminate, a drug development program. Clinical detection of DIVI can be very difficult as there are no definitive biomarkers known to reliably detect this disorder in all instances. The preclinical identification of DIVI requires detailed microscopic examination of a wide range of tissues although one of the most commonly affected areas in rats is the mesenteric vasculature. The reason for this predisposition of mesenteric arteries in rats as well as the exact mechanism and cell types involved in the initial development of these lesions have not been fully elucidated. We hypothesized that by using a mixed culture of cells from rat mesenteric tissue, we would be able to identify an RNA expression signature that could predict the invivo development of DIVI. Five compounds designed to inhibit Phosphodiesterase 4 activity (PDE4i) were chosen as positive controls. PDE4i's are well known to induce DIVI in the mesenteric vasculature of rats and there is microscopic evidence that this is associated, at least in part, with a proinflammatory mechanism. We surveyed, by qRT-PCR, the expression of 96 genes known to be involved in inflammation and using a Random-Forest model, identified 12 genes predictive of invivo DIVI outcomes in rats. Using these genes, we were able to cross-validate the ability of the Random-Forest modeling to predict the concentration at which PDE4i caused DIVI invivo.


Sujet(s)
Artères mésentériques/cytologie , Inhibiteurs de la phosphodiestérase-4/toxicité , Lésions du système vasculaire/induit chimiquement , Animaux , Mâle , Rats , Rat Sprague-Dawley
7.
Toxicol Sci ; 155(1): 148-156, 2017 01.
Article de Anglais | MEDLINE | ID: mdl-27708194

RÉSUMÉ

Tofacitinib is an oral Janus kinase (JAK) inhibitor for the treatment of rheumatoid arthritis. Tofacitinib preferentially inhibits receptor signaling through JAK3 and JAK1, relative to JAK2. In the 2-year rat carcinogenicity study, there were tofacitinib, dose-related increases in the incidences of testicular Leydig cell hyperplasia and benign adenomas in male rats, and decreased incidences of mammary tumors and duct dilatation/galactocele in female rats. Such findings in rats are typical of agents, such as dopamine agonists, which decrease prolactin (PRL) activity. Since prolactin signals through the JAK2 pathway, we hypothesized that these findings were off-target effects due to inhibition of PRL signaling via JAK2. The studies reported here were designed to investigate the interruption of PRL signaling pathways in Leydig cells. In isolated primary rat Leydig cells, PRL increased phosphorylated Signal Transducer and Activator of Transcription-5 protein, and mRNA levels for luteinizing hormone receptor. Tofacitinib, at concentrations observed in the rat carcinogenicity study, dose-dependently inhibited these effects. These observations illustrate a novel mechanism, the inhibition of prolactin signaling by which modulation of JAK activity can modulate PRL signaling pathways to induce Leydig cell tumors in rats. Since human Leydig cells lack this PRL dependence for normal function, these rodent tumors do not indicate a health risk to human patients.


Sujet(s)
Adénomes/anatomopathologie , Hyperplasie/induit chimiquement , Cellules de Leydig/effets des médicaments et des substances chimiques , Pipéridines/pharmacologie , Prolactine/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Pyrimidines/pharmacologie , Pyrroles/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Testicule/effets des médicaments et des substances chimiques , Animaux , Cellules de Leydig/métabolisme , Mâle , Rats , Testicule/anatomopathologie
8.
Toxicol Sci ; 138(1): 234-48, 2014 Mar.
Article de Anglais | MEDLINE | ID: mdl-24336348

RÉSUMÉ

Development of LNA gapmers, antisense oligonucleotides used for efficient inhibition of target RNA expression, is limited by non-target mediated hepatotoxicity issues. In the present study, we investigated hepatic transcription profiles of mice administered non-toxic and toxic LNA gapmers. After repeated administration, a toxic LNA gapmer (TS-2), but not a non-toxic LNA gapmer (NTS-1), caused hepatocyte necrosis and increased serum alanine aminotransferase levels. Microarray data revealed that, in addition to gene expression patterns consistent with hepatotoxicity, 17 genes in the clathrin-mediated endocytosis (CME) pathway were altered in the TS-2 group. TS-2 significantly down-regulated myosin 1E (Myo1E), which is involved in release of clathrin-coated pits from plasma membranes. To map the earliest transcription changes associated with LNA gapmer-induced hepatotoxicity, a second microarray analysis was performed using NTS-1, TS-2, and a severely toxic LNA gapmer (HTS-3) at 8, 16, and 72 h following a single administration in mice. The only histopathological change observed was minor hepatic hypertrophy in all LNA groups across time points. NTS-1, but not 2 toxic LNA gapmers, increased immune response genes at 8 and 16 h but not at 72 h. TS-2 significantly perturbed the CME pathway only at 72 h, while Myo1E levels were decreased at all time points. In contrast, HTS-3 modulated DNA damage pathway genes at 8 and 16 h and also modulated the CME pathway genes (but not Myo1E) at 16 h. Our results may suggest that different LNAs modulate distinct transcriptional genes and pathways contributing to non-target mediated hepatotoxicity in mice.


Sujet(s)
Lésions hépatiques dues aux substances/anatomopathologie , Endocytose/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Oligonucléotides antisens/toxicité , Oligonucléotides/toxicité , Transcriptome/effets des médicaments et des substances chimiques , Animaux , Lésions hépatiques dues aux substances/étiologie , Lésions hépatiques dues aux substances/génétique , Clathrine/métabolisme , Endocytose/génétique , Analyse de profil d'expression de gènes , Injections sous-cutanées , Foie/anatomopathologie , Mâle , Souris , Lignées consanguines de souris , Données de séquences moléculaires , Oligonucléotides/composition chimique , Oligonucléotides/génétique , Oligonucléotides/métabolisme , Oligonucléotides antisens/composition chimique , Oligonucléotides antisens/génétique , Oligonucléotides antisens/métabolisme
9.
Toxicol Appl Pharmacol ; 274(1): 156-67, 2014 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-24126418

RÉSUMÉ

Pretreatment of mice with a low hepatotoxic dose of acetaminophen (APAP) results in resistance to a subsequent, higher dose of APAP. This mouse model, termed APAP autoprotection was used here to identify differentially expressed genes and cellular pathways that could contribute to this development of resistance to hepatotoxicity. Male C57BL/6J mice were pretreated with APAP (400mg/kg) and then challenged 48h later with 600mg APAP/kg. Livers were obtained 4 or 24h later and total hepatic RNA was isolated and hybridized to Affymetrix Mouse Genome MU430_2 GeneChip. Statistically significant genes were determined and gene expression changes were also interrogated using the Causal Reasoning Engine (CRE). Extensive literature review narrowed our focus to methionine adenosyl transferase-1 alpha (MAT1A), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), flavin-containing monooxygenase 3 (Fmo3) and galectin-3 (Lgals3). Down-regulation of MAT1A could lead to decreases in S-adenosylmethionine (SAMe), which is known to protect against APAP toxicity. Nrf2 activation is expected to play a role in protective adaptation. Up-regulation of Lgals3, one of the genes supporting the Nrf2 hypothesis, can lead to suppression of apoptosis and reduced mitochondrial dysfunction. Fmo3 induction suggests the involvement of an enzyme not known to metabolize APAP in the development of tolerance to APAP toxicity. Subsequent quantitative RT-PCR and immunochemical analysis confirmed the differential expression of some of these genes in the APAP autoprotection model. In conclusion, our genomics strategy identified cellular pathways that might further explain the molecular basis for APAP autoprotection.


Sujet(s)
Acétaminophène/toxicité , Lésions hépatiques dues aux substances/métabolisme , Lésions hépatiques dues aux substances/anatomopathologie , Régulation de l'expression des gènes/physiologie , Modèles animaux , Animaux , Lésions hépatiques dues aux substances/génétique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Mâle , Souris , Souris de lignée C57BL
10.
Toxicol Sci ; 128(1): 42-56, 2012 Jul.
Article de Anglais | MEDLINE | ID: mdl-22539613

RÉSUMÉ

The preceding article identified key components of pregabalin's mode of action on nongenotoxic hemangiosarcoma formation in mice, including increased serum bicarbonate leading to decreased respiratory rate, increased blood pH, increased venous oxygen saturation, increased vascular endothelial growth factor and basic fibroblast growth factor expression, increased hepatic vascular endothelial growth factor receptor 2 expression, and increased iron-laden macrophages. Increased platelet count and platelet activation were early, species-specific biomarkers in mice. Dysregulated erythropoiesis, macrophage activation, and elevations of tissue growth factors were consistent with the unified mode of action for nongenotoxic hemangiosarcoma recently proposed at an international hemangiosarcoma workshop (Cohen, S. M., Storer, R. D., Criswell, K. A., Doerrer, N. G., Dellarco, V. L., Pegg, D. G., Wojcinski, Z. W., Malarkey, D. E., Jacobs, A. C., Klaunig, J. E., et al. (2009). Hemangiosarcoma in rodents: Mode-of-action evaluation and human relevance. Toxicol. Sci. 111, 4-18). In this article, we present evidence that pregabalin induces hypoxia and increases endothelial cell (EC) proliferation in a species-specific manner. Dietary administration of pregabalin produced a significant 35% increase in an immunohistochemical stain for hypoxia (Hypoxyprobe) in livers from pregabalin-treated mice. Increased Hypoxyprobe staining was not observed in the liver, bone marrow, or spleen of rats, supporting the hypothesis that pregabalin produces local tissue hypoxia in a species-specific manner. Transcriptional analysis supports that rats, unlike mice, adapt to pregabalin-induced hypoxia. Using a dual-label method, increased EC proliferation was observed as early as 2 weeks in mouse liver and 12 weeks in bone marrow following pregabalin administration. These same assays showed decreased EC proliferation in hepatic ECs of rats, further supporting species specificity. Dietary supplementation with vitamin E, which is known to have antioxidant and antiangiogenic activity, inhibited pregabalin-induced increases in mouse hepatic EC proliferation, providing confirmatory evidence for the proposed mode of action and its species-specific response.


Sujet(s)
Hypoxie cellulaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Endothélium vasculaire/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Vitamine E/administration et posologie , Acide gamma-amino-butyrique/analogues et dérivés , Animaux , Chromatographie en phase liquide à haute performance , Régime alimentaire , Endothélium vasculaire/cytologie , Femelle , Foie/cytologie , Souris , Prégabaline , Acide gamma-amino-butyrique/toxicité
11.
Toxicol Pathol ; 39(7): 1032-45, 2011 Dec.
Article de Anglais | MEDLINE | ID: mdl-21937740

RÉSUMÉ

Peroxisome proliferator-activated receptor gamma (PPARγ) agonists and PPARγ/α dual agonists are used in the treatment of type 2 diabetes mellitus and hyperlipidemias. In carcinogenicity studies, some of these agonists induced hemangiomas/hemangiosarcomas in mice, but not in rats. We hypothesized that increased endothelial cell (EC) proliferation may be involved in the mechanism of PPAR agonist-induced vascular tumors in mice. We previously showed that the sarcomagenic PPARγ agonist troglitazone (TG) increased EC proliferation in brown and white adipose tissue and liver in mice at sarcomagenic doses (400 and 800 mg/kg) after four weeks of treatment. In vitro, TG had a mitogenic effect on mouse microvascular mouse ECs by increasing cell proliferation and survival. The current studies showed that treatment of mouse ECs in vitro induced alterations in proliferation pathway gene expression, especially the expression of insulin-like growth factor-1, but had no effect on mouse oxidative stress pathways. In vivo, treatment with vitamin E did not inhibit TG-induced EC proliferation in liver and adipose tissue. In addition, no hypoxic effect was detected in adipose tissue of TG-treated mice; however, TG had a minor effect on hepatocellular hypoxia. These results provide additional evidence supporting a direct mitogenic effect in the mode of action of TG-induced hemangiosarcomas in mice.


Sujet(s)
Processus de croissance cellulaire/effets des médicaments et des substances chimiques , Chromanes/pharmacologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Récepteur PPAR gamma/agonistes , Thiazolidinediones/pharmacologie , Tissu adipeux/composition chimique , Animaux , Antinéoplasiques/pharmacologie , Poids/effets des médicaments et des substances chimiques , Hypoxie cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Consommation de boisson/effets des médicaments et des substances chimiques , Cellules endothéliales/cytologie , Cellules endothéliales/métabolisme , Stéatose hépatique , Femelle , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Réseaux de régulation génique , Hémangiosarcome , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Histocytochimie , Facteur de croissance IGF-I/génétique , Facteur de croissance IGF-I/métabolisme , Foie/composition chimique , Foie/effets des médicaments et des substances chimiques , Souris , Taille d'organe/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Reproductibilité des résultats , Troglitazone , Vitamine E/pharmacologie
12.
Am J Pathol ; 179(1): 240-7, 2011 Jul.
Article de Anglais | MEDLINE | ID: mdl-21703406

RÉSUMÉ

Macrophage colony-stimulating factor (M-CSF) is a hematopoietic growth factor that is responsible for the survival and proliferation of monocytes and the differentiation of monocytes into macrophages, including Kupffer cells (KCs) in the liver. KCs play an important role in the clearance of several serum enzymes, including aspartate aminotransferase and creatine kinase, that are typically elevated as a result of liver or skeletal muscle injury. We used three distinct animal models to investigate the hypothesis that increases in the levels of serum enzymes can be the result of decreases in KCs in the apparent absence of hepatic or skeletal muscle injury. Specifically, neutralizing M-CSF activity via a novel human monoclonal antibody reduced the CD14(+)CD16(+) monocyte population, depleted KCs, and increased aspartate aminotransferase and creatine kinase serum enzyme levels in cynomolgus macaques. In addition, the treatment of rats with clodronate liposomes depleted KCs and led to increased serum enzyme levels, again without evidence of tissue injury. Finally, in the osteopetrotic (Csf1(op)/Csf1(op)) mice lacking functional M-CSF and having reduced levels of KCs, the levels of serum enzymes are higher than in wild-type littermates. Together, these findings support a mechanism for increases in serum enzyme levels through M-CSF regulation of tissue macrophage homeostasis without concomitant histopathological changes in either the hepatic or skeletal system.


Sujet(s)
Aspartate aminotransferases/sang , Creatine kinase/sang , Cellules de Küpffer/anatomopathologie , Foie/enzymologie , Muscles squelettiques/enzymologie , Ostéopétrose/anatomopathologie , Animaux , Anticorps monoclonaux/pharmacologie , Agents de maintien de la densité osseuse/pharmacologie , Acide clodronique/pharmacologie , Test ELISA , Femelle , Humains , Cellules de Küpffer/effets des médicaments et des substances chimiques , Cellules de Küpffer/métabolisme , Antigènes CD14/métabolisme , Foie/traumatismes , Foie/anatomopathologie , Macaca fascicularis , Facteur de stimulation des colonies de macrophages/métabolisme , Facteur de stimulation des colonies de macrophages/physiologie , Mâle , Souris , Souris knockout , Monocytes/effets des médicaments et des substances chimiques , Monocytes/métabolisme , Monocytes/anatomopathologie , Muscles squelettiques/traumatismes , Muscles squelettiques/anatomopathologie , Ostéopétrose/métabolisme , Rats , Rat Sprague-Dawley , Récepteurs du fragment Fc des IgG/métabolisme
13.
Toxicol Sci ; 113(1): 254-66, 2010 Jan.
Article de Anglais | MEDLINE | ID: mdl-19812364

RÉSUMÉ

To understand the molecular mechanisms underlying compound-induced hemangiosarcomas in mice, and therefore, their human relevance, a systems biology approach was undertaken using transcriptomics and Causal Network Modeling from mice treated with 2-butoxyethanol (2-BE). 2-BE is a hemolytic agent that induces hemangiosarcomas in mice. We hypothesized that the hemolysis induced by 2-BE would result in local tissue hypoxia, a well-documented trigger for endothelial cell proliferation leading to hemangiosarcoma. Gene expression data from bone marrow (BM), liver, and spleen of mice exposed to a single dose (4 h) or seven daily doses of 2-BE were used to develop a mechanistic model of hemangiosarcoma. The resulting mechanistic model confirms previous work proposing that 2-BE induces macrophage activation and inflammation in the liver. In addition, the model supports local tissue hypoxia in the liver and spleen, coupled with increased erythropoeitin signaling and erythropoiesis in the spleen and BM, and suppression of mechanisms that contribute to genomic stability, events that could be contributing factors to hemangiosarcoma formation. Finally, an immunohistochemistry method (Hypoxyprobe) demonstrated that tissue hypoxia was present in the spleen and BM. Together, the results of this study identify molecular mechanisms that initiate hemangiosarcoma, a key step in understanding safety concerns that can impact drug decision processes, and identified hypoxia as a possible contributing factor for 2-BE-induced hemangiosarcoma in mice.


Sujet(s)
Moelle osseuse/métabolisme , Transformation cellulaire néoplasique/métabolisme , Hémangiosarcome/métabolisme , Foie/métabolisme , Modèles biologiques , Transduction du signal , Rate/métabolisme , Biologie des systèmes , Animaux , Moelle osseuse/anatomopathologie , Cycle cellulaire , Différenciation cellulaire , Hypoxie cellulaire , Prolifération cellulaire , Transformation cellulaire néoplasique/induit chimiquement , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/anatomopathologie , Modèles animaux de maladie humaine , Cellules endothéliales/métabolisme , Érythropoïèse , Érythropoïétine/métabolisme , Éthylène glycols , Analyse de profil d'expression de gènes/méthodes , Régulation de l'expression des gènes tumoraux , Instabilité du génome , Hémangiosarcome/induit chimiquement , Hémangiosarcome/génétique , Hémangiosarcome/anatomopathologie , Cellules souches hématopoïétiques/métabolisme , Hémolyse , Hépatite/métabolisme , Hépatite/anatomopathologie , Immunohistochimie , Foie/anatomopathologie , Activation des macrophages , Mâle , Souris , Rate/anatomopathologie , Facteurs temps
14.
Toxicol Appl Pharmacol ; 222(2): 169-79, 2007 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-17585979

RÉSUMÉ

Pretreatment of mice with the peroxisome proliferator clofibrate (CFB) protects against acetaminophen (APAP)-induced hepatotoxicity. Previous studies have shown that activation of the nuclear peroxisome proliferator activated receptor-alpha (PPARalpha) is required for this effect. The present study utilizes gene expression profile analysis to identify potential pathways contributing to PPARalpha-mediated hepatoprotection. Gene expression profiles were compared between wild type and PPARalpha-null mice pretreated with vehicle or CFB (500 mg/kg, i.p., daily for 10 days) and then challenged with APAP (400 mg/kg, p.o.). Total hepatic RNA was isolated 4 h after APAP treatment and hybridized to Affymetrix Mouse Genome MGU74 v2.0 GeneChips. Gene expression analysis was performed utilizing GeneSpring software. Our analysis identified 53 genes of interest including vanin-1, cell cycle regulators, lipid-metabolizing enzymes, and aldehyde dehydrogenase 2, an acetaminophen binding protein. Vanin-1 could be important for CFB-mediated hepatoprotection because this protein is involved in the synthesis of cysteamine and cystamine. These are potent antioxidants capable of ameliorating APAP toxicity in rodents and humans. HPLC-ESI/MS/MS analysis of liver extracts indicates that enhanced vanin-1 gene expression results in elevated cystamine levels, which could be mechanistically associated with CFB-mediated hepatoprotection.


Sujet(s)
Clofibrate/pharmacologie , Analyse de profil d'expression de gènes/méthodes , Foie/effets des médicaments et des substances chimiques , Récepteur PPAR alpha/génétique , Acétaminophène/administration et posologie , Acétaminophène/toxicité , Acyl-CoA oxidase/génétique , Acyl-CoA oxidase/métabolisme , Amidohydrolases , Animaux , Anticholestérolémiants/pharmacologie , Anticholestérolémiants/usage thérapeutique , Molécules d'adhérence cellulaire/génétique , Molécules d'adhérence cellulaire/métabolisme , Lésions hépatiques dues aux substances , Clofibrate/usage thérapeutique , Analyse de regroupements , Cystamine/composition chimique , Cystamine/métabolisme , Mercaptamine/composition chimique , Mercaptamine/métabolisme , Énoyl-CoA hydratases/génétique , Énoyl-CoA hydratases/métabolisme , Protéines liées au GPI , Foie/métabolisme , Foie/anatomopathologie , Maladies du foie/génétique , Maladies du foie/prévention et contrôle , Malate dehydrogenase/génétique , Malate dehydrogenase/métabolisme , Mâle , Souris , Lignées consanguines de souris , Souris knockout , Séquençage par oligonucléotides en batterie/méthodes , Récepteur PPAR alpha/métabolisme , Pantéthéine/composition chimique , Pantéthéine/métabolisme , Acide pantothénique/composition chimique , Acide pantothénique/métabolisme , Proliférateurs des péroxysomes/métabolisme , Proteasome endopeptidase complex/génétique , Proteasome endopeptidase complex/métabolisme , RT-PCR
15.
Cancer Res ; 64(21): 8109-15, 2004 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-15520223

RÉSUMÉ

Retinoids, natural and synthetic derivatives of vitamin A, are active in cancer therapy and chemoprevention. We reported previously that all-trans-retinoic acid (RA) treatment prevented carcinogen-induced transformation of immortalized human bronchial epithelial (HBE) cells. To identify cancer chemopreventive mechanisms, immortalized (BEAS-2B), carcinogen-transformed (BEAS-2B(NNK)), and RA-chemoprevented (BEAS-2B(NNK/RA)) HBE cells were used to conduct microarray analyses independently. Species increased in chemoprevented as compared with immortalized HBE cells (group I) and those augmented in chemoprevented as compared with transformed HBE cells (group II) included known RA-target genes as well as previously unrecognized RA-target genes in HBE cells. Unexpectedly, both groups were also enriched for interferon-stimulated genes. One interferon-stimulated gene of particular interest was UBE1L, the ubiquitin-activating enzyme E1-like protein. UBE1L expression was also induced after prolonged RA-treatment of immortalized HBE cells. UBE1L mRNA was shown previously as repressed in certain lung cancer cell lines, directly implicating UBE1L in lung carcinogenesis. Notably, UBE1L immunoblot expression was reduced in a subset of malignant as compared with adjacent normal lung tissues that were examined. Immunohistochemical analyses were performed using a new assay developed to detect this species using rabbit polyclonal anti-UBE1L antibodies independently raised against the amino- or carboxyl-termini of UBE1L. Studies done on paraffin-embedded and fixed tissues revealed abundant UBE1L, but low levels of cyclin D1 expression in the normal human bronchial epithelium, indicating an inverse relationship existed between these species. To study this further, cotransfection into HBE cells of wild-type or mutant UBE1L species was accomplished. In a dose-dependent manner, wild-type but not mutant UBE1L species repressed cyclin D1 expression. This implicated UBE1L in a retinoid chemoprevention mechanism involving cyclin D1 repression described previously. Taken together, these findings directly implicate UBE1L as a candidate-pharmacologic target for lung cancer chemoprevention. These findings also provide a mechanistic basis for the tumor suppressive effects of UBE1L through cyclin D1 repression.


Sujet(s)
Anticarcinogènes/usage thérapeutique , Tumeurs du poumon/prévention et contrôle , Trétinoïne/usage thérapeutique , Ubiquitin-activating enzymes/génétique , Lignée cellulaire tumorale , Gènes suppresseurs de tumeur , Humains , Immunohistochimie , Tumeurs du poumon/génétique , Séquençage par oligonucléotides en batterie , Ubiquitin-activating enzymes/analyse , Ubiquitin-activating enzymes/physiologie
16.
EMBO J ; 22(24): 6526-36, 2003 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-14657025

RÉSUMÉ

Nur77 is a nuclear orphan steroid receptor that has been implicated in negative selection. Expression of Nur77 in thymocytes and cell lines leads to apoptosis through a mechanism that remains unclear. In some cell lines, Nur77 was reported to act through a transcription-independent mechanism involving translocation to mitochondria, leading to cytochrome c release. However, we show here that Nur77-mediated apoptosis in thymocytes does not involve cytoplasmic cytochrome c release and cannot be rescued by Bcl-2. Microarray analysis shows that Nur77 induces many genes, including two novel genes (NDG1, NDG2) and known apoptotic genes FasL and TRAIL. Characterization of NDG1 and NDG2 indicates that NDG1 initiates a novel apoptotic pathway in a Bcl-2-independent manner. Thus Nur77-mediated apoptosis in T cells involves Bcl-2 independent transcriptional activation of several known and novel apoptotic pathways.


Sujet(s)
Apoptose/physiologie , Protéines de liaison à l'ADN/physiologie , Récepteurs aux stéroïdes/physiologie , Lymphocytes T/physiologie , Facteurs de transcription/physiologie , Activation de la transcription/génétique , Séquence d'acides aminés , Animaux , Séquence nucléotidique , Cellules cultivées , Clonage moléculaire , Amorces ADN , Protéines de liaison à l'ADN/génétique , Escherichia coli/cytologie , Escherichia coli/génétique , Escherichia coli/physiologie , Femelle , Génotype , Humains , Souris , Souris de lignée C57BL , Souris transgéniques , Données de séquences moléculaires , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Séquençage par oligonucléotides en batterie , Grossesse , Rats , Récepteurs aux antigènes des cellules T/physiologie , Récepteurs cytoplasmiques et nucléaires , RT-PCR , Lymphocytes T/cytologie , Lymphocytes T/immunologie , Facteurs de transcription/génétique
17.
Oncogene ; 22(31): 4924-32, 2003 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-12894236

RÉSUMÉ

Retinoids, the natural and synthetic derivatives of vitamin A, have a role in cancer treatment and prevention. There is a need to reveal mechanisms that account for retinoid response or resistance. This study identified candidate all-trans-retinoic acid (RA) target genes linked to growth suppression in BEAS-2B human bronchial epithelial cells. Microarray analyses were performed using Affymetrix arrays. A total of 11 RA-induced species were validated by reverse transcription polymerase chain reaction (RT-PCR), Western or Northern analyses. Three of these species were novel candidate RA-target genes in human bronchial epithelial cells. These included: placental bone morphogenetic protein (PLAB), polyamine oxidase isoform 1 (PAOh1) and E74-like factor 3 (ELF3). Expression patterns were studied in RA-resistant BEAS-2B-R1 cells. In BEAS-2B-R1 cells, RA dysregulated the expression of the putative lymphocyte G0/G1 switch gene (G0S2), heme oxygenase 1 (HMOX1), tumor necrosis factor-alpha-induced protein 2 (TNFAIP2), inhibitor of DNA binding 1(Id1), fos-like antigen 1 (FOSL1), transglutaminase 2 (TGM2), asparagine synthetase (ASNS), PLAB, PAOh1 and ELF3, while prominent induction of insulin-like growth-factor-binding protein 6 (IGFBP6) still occurred. In summary, this study identified 11 candidate RA-target genes in human bronchial epithelial cells including three novel species. Expression studies in BEAS-2B-R1 cells indicated that several were directly implicated in RA signaling, since their aberrant expression was linked to RA resistance of human bronchial epithelial cells.


Sujet(s)
Bronches/cytologie , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Trétinoïne/pharmacologie , Technique de Northern , Technique de Western , Lignée de cellules transformées/effets des médicaments et des substances chimiques , Lignée de cellules transformées/métabolisme , Milieux de culture sans sérum/pharmacologie , Résistance aux substances , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Humains , Séquençage par oligonucléotides en batterie , ARN messager/biosynthèse , ARN messager/génétique , ARN tumoral/biosynthèse , ARN tumoral/génétique , RT-PCR
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...