Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 9 de 9
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Redox Biol ; 71: 103074, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38367511

RÉSUMÉ

Brain iron accumulation constitutes a pathognomonic indicator in several neurodegenerative disorders. Metal accumulation associated with dopaminergic neuronal death has been documented in Parkinson's disease. Through the use of in vivo and in vitro models, we demonstrated that lipid dysregulation manifests as a neuronal and glial response during iron overload. In this study, we show that cholesterol content and triacylglycerol (TAG) hydrolysis were strongly elevated in mice midbrain. Lipid cacostasis was concomitant with the loss of dopaminergic neurons, astrogliosis and elevated expression of α-synuclein. Exacerbated lipid peroxidation and markers of ferroptosis were evident in the midbrain from mice challenged with iron overload. An imbalance in the activity of lipolytic and acylation enzymes was identified, favoring neutral lipid hydrolysis, and consequently reducing TAG and cholesteryl ester levels. Notably, these observed alterations were accompanied by motor impairment in iron-treated mice. In addition, neuronal and glial cultures along with their secretomes were used to gain further insight into the mechanism underlying TAG hydrolysis and cholesterol accumulation as cellular responses to iron accumulation. We demonstrated that TAG hydrolysis in neurons is triggered by astrocyte secretomes. Moreover, we found that the ferroptosis inhibitor, ferrostatin-1, effectively prevents cholesterol accumulation both in neurons and astrocytes. Taken together, these results indicate that lipid disturbances occur in iron-overloaded mice as a consequence of iron-induced oxidative stress and depend on neuron-glia crosstalk. Our findings suggest that developing therapies aimed at restoring lipid homeostasis may lead to specific treatment for neurodegeneration associated with ferroptosis and brain iron accumulation.


Sujet(s)
Ferroptose , Surcharge en fer , Troubles moteurs , Souris , Animaux , Métabolisme lipidique , Troubles moteurs/métabolisme , Fer/métabolisme , Peroxydation lipidique , Neurones dopaminergiques/métabolisme , Cholestérol/métabolisme , Lipides
2.
J Steroid Biochem Mol Biol ; 154: 285-93, 2015 Nov.
Article de Anglais | MEDLINE | ID: mdl-26365558

RÉSUMÉ

The active form of vitamin D3, calcitriol, plays a major role in maintaining calcium/phosphate homeostasis. In addition, it is a potent antiproliferative and prodifferentiating agent. However, when effective antitumor doses of calcitriol are employed, hypercalcemic effects are observed, thus precluding its therapeutic application. To overcome this problem, structural analogues have been designed with the aim at retaining or even increasing the antitumor effects while decreasing its calcemic activity. This report shows the biological evaluation of an alkynylphosphonate vitamin D less-calcemic analogue in a murine model of breast cancer. We demonstrate that this compound has potent anti-metastatic effects through its action over cellular migration and invasion likely mediated through the up-regulation of E-cadherin expression. Based on the current in vitro and in vivo results, EM1 is a promising candidate as a therapeutic agent in breast cancer.


Sujet(s)
Tumeurs du sein/anatomopathologie , Calcitriol/pharmacologie , Métastase tumorale/prévention et contrôle , Phosphonates/pharmacologie , Animaux , Calcitriol/analogues et dérivés , Modèles animaux de maladie humaine , Femelle , Souris , Souris de lignée BALB C
3.
Exp Mol Pathol ; 97(3): 411-24, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25240203

RÉSUMÉ

There is evidence that p300, a transcriptional co-factor and a lysine acetyl-transferase, could play a role both as an oncoprotein and as a tumor suppressor, although little is known regarding its role in breast cancer (BC). First we investigated the role p300 has on BC by performing pharmacological inhibition of p300 acetyl-transferase function and analyzing the effects on cell count, migration and invasion in LM3 murine breast cancer cell line and on tumor progression in a syngeneic murine model. We subsequently studied p300 protein expression in human BC biopsies and evaluated its correlation with clinical and histopathological parameters of the patients. We observed that inhibition of p300 induced apoptosis and reduced migration and invasion in cultured LM3 cells. Furthermore, a significant reduction in tumor burden, number of lung metastases and number of tumors invading the abdominal cavity was observed in a syngeneic tumor model of LM3 following treatment with the p300 inhibitor. This reduction in tumor burden was accompanied by a decrease in the mitotic index and Ki-67 levels and an increase in Bax expression. Moreover, the analysis of p300 expression in human BC samples showed that p300 immunoreactivity is significantly higher in the cancerous tissues than in the non-malignant mammary tissues and in the histologically normal adjacent tissues. Interestingly, p300 was observed in the cytoplasm, and the rate of cytoplasmic p300 was higher in BC than in non-tumor tissues. Importantly, we found that cytoplasmic localization of p300 is associated with a longer overall survival time of the patients. In conclusion, we demonstrated that inhibition of the acetylase function of p300 reduces both cell count and invasion in LM3 cells, and decreases tumor progression in the animal model. In addition, we show that the presence of p300 in the cytoplasm correlates with increased survival of patients suggesting that its nuclear localization is necessary for the pro-tumoral effects.


Sujet(s)
Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Protéine p300-E1A/métabolisme , Animaux , Apoptose/physiologie , Technique de Western , Tumeurs du sein/mortalité , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire/physiologie , Cytoplasme/composition chimique , Cytoplasme/métabolisme , Modèles animaux de maladie humaine , Femelle , Technique d'immunofluorescence , Humains , Estimation de Kaplan-Meier , Souris , Souris de lignée BALB C , Microscopie confocale
4.
J Neurooncol ; 118(1): 49-60, 2014 May.
Article de Anglais | MEDLINE | ID: mdl-24584679

RÉSUMÉ

Vitamin D and its analogs have been shown to display anti-proliferative effects in a wide variety of cancer types including glioblastoma multiforme (GBM). These anticancer effects are mediated by its active metabolite, 1α, 25-dihydroxyvitamin D3 (calcitriol) acting mainly through vitamin D receptor (VDR) signaling. In addition to its involvement in calcitriol action, VDR has also been demonstrated to be useful as a prognostic factor for some types of cancer. However, to our knowledge, there are no studies evaluating the expression of VDR protein and its association with outcome in gliomas. Therefore, we investigated VDR expression by using immunohistochemical analysis in human glioma tissue microarrays, and analyzed the association between VDR expression and clinico-pathological parameters. We further investigated the effects of genetic and pharmacologic modulation of VDR on survival and migration of glioma cell lines. Our data demonstrate that VDR is increased in tumor tissues when compared with VDR in non-malignant brains, and that VDR expression is associated with an improved outcome in patients with GBM. We also show that both genetic and pharmacologic modulation of VDR modulates GBM cellular migration and survival and that VDR is necessary for calcitriol-mediated effects on migration. Altogether these results provide some limited evidence supporting a role for VDR in glioma progression.


Sujet(s)
Tumeurs du cerveau/métabolisme , Régulation de l'expression des gènes tumoraux/génétique , Glioblastome/métabolisme , Récepteur calcitriol/métabolisme , Adulte , Facteurs âges , Tumeurs du cerveau/mortalité , Tumeurs du cerveau/anatomopathologie , Calcitriol/pharmacologie , Agonistes des canaux calciques/pharmacologie , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/physiologie , Cycline D1/métabolisme , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Glioblastome/mortalité , Glioblastome/anatomopathologie , Humains , Mâle , Adulte d'âge moyen , Protéines oncogènes/métabolisme , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Facteurs sexuels , Facteurs temps , Analyse sur puce à tissus
5.
Tumour Biol ; 35(3): 2803-15, 2014 Mar.
Article de Anglais | MEDLINE | ID: mdl-24234335

RÉSUMÉ

In human glioma tumors, heme oxygenase-1 (HO-1) has been shown to be upregulated both when compared with normal brain tissues and also during oligodendroglioma progression. The cell types that express HO-1 have been shown to be mainly macrophages/microglia and T cells. However, many other reports also demonstrated that cell lines derived from glioma tumors and astrocytes express HO-1 after the occurrence of a wide variety of cell injuries and stressors. In addition, the significance of HO-1 upregulation in glioma had not, so far, been addressed. We therefore aimed at investigating the expression and significance of HO-1 in human glial tumors. For this purpose, we performed a wide screening of HO-1 expression in gliomas by using tissue microarrays containing astrocytomas, oligodendrogliomas, mixed tumors, and normal brain tissues. We subsequently correlated protein expression with patient clinicopathological data. We found differences in HO-1 positivity rates between non-malignant brain (22 %) and gliomas (54%, p = 0.01). HO-1 was expressed by tumor cells and showed cytoplasmic localization, although 19% of tumor samples also depicted nuclear staining. Importantly, a significant decrease in the overall survival time of grade II and III astrocytoma patients with HO-1 expression was observed. This result was validated at the mRNA level in a cohort of 105 samples. However, no association of HO-1 nuclear localization with patient survival was detected. In vitro experiments aimed at investigating the role of HO-1 in glioma progression showed that HO-1 modulates glioma cell proliferation, but has no effects on cellular migration. In conclusion, our results corroborate the higher frequency of HO-1 protein expression in gliomas than in normal brain, demonstrate that HO-1 is expressed by glial malignant cells, and show an association of HO-1 expression with patients' shorter survival time.


Sujet(s)
Marqueurs biologiques tumoraux/analyse , Tumeurs du cerveau/enzymologie , Gliome/enzymologie , Heme oxygenase-1/biosynthèse , Astrocytome/enzymologie , Astrocytome/mortalité , Astrocytome/anatomopathologie , Tumeurs du cerveau/mortalité , Tumeurs du cerveau/anatomopathologie , Technique d'immunofluorescence , Gliome/mortalité , Gliome/anatomopathologie , Heme oxygenase-1/analyse , Humains , Immunotransfert , Immunohistochimie , Estimation de Kaplan-Meier , Pronostic , Réaction de polymérisation en chaine en temps réel , Analyse sur puce à tissus
7.
Exp Mol Pathol ; 93(2): 237-45, 2012 Oct.
Article de Anglais | MEDLINE | ID: mdl-22580187

RÉSUMÉ

The expression of heme oxygenase-1 (HO-1) was shown to be increased in multiple tumors compared with their surrounding healthy tissues and was also observed to be up-regulated in oral squamous cell carcinomas (OSCC). However, conflicting results were obtained and little information is available regarding HO-1 significance in head and neck squamous cell carcinoma (HNSCC). Therefore, the aim of the present study was to perform a wide screening of HO-1 expression in a large collection of human primary HNSCCs and to correlate the results with clinical and pathological parameters. For this purpose, we investigated the expression of this protein by immunohistochemistry (IHC) in tissue microarrays (TMAs) of HNSCC and in an independent cohort of paraffin-embedded tumor specimens. HO-1 expression was further validated by real-time qPCR performed on selected laser capture-microdissected (LCM) oral tissue samples. Both the number of HO-1-positive samples and HO-1 immunoreactivity in the cancerous tissues were significantly higher than those in the non-tumor tissues. These results were confirmed at the mRNA level. Interestingly, HO-1 localization was observed in the nucleus, and the rate of nuclear HO-1 in HNSCC was higher than that in non-malignant tissues. Nuclear HO-1 was observed in HNSCC cell lines and increased even further following hemin treatment. Analysis of HO-1 expression and sub-cellular localization in a mouse model of squamous cell carcinoma (SCC) and in human HNSCC revealed that nuclear HO-1 increases with tumor progression. Taken together, these results demonstrate that HO-1 is up-regulated in HNSCC and that nuclear localization of HO-1 is associated with malignant progression in this tumor type.


Sujet(s)
Carcinome épidermoïde/enzymologie , Tumeurs de la tête et du cou/enzymologie , Heme oxygenase-1/métabolisme , Sujet âgé , Animaux , Carcinome épidermoïde/diagnostic , Carcinome épidermoïde/génétique , Modèles animaux de maladie humaine , Évolution de la maladie , Femelle , Tumeurs de la tête et du cou/diagnostic , Tumeurs de la tête et du cou/génétique , Heme oxygenase-1/génétique , Humains , Mâle , Souris , Adulte d'âge moyen , Inclusion en paraffine , Réaction de polymérisation en chaîne , Pronostic , ARN messager/métabolisme , Analyse sur puce à tissus
8.
Cells Tissues Organs ; 192(5): 314-24, 2010.
Article de Anglais | MEDLINE | ID: mdl-20606403

RÉSUMÉ

Sphingosine kinase-1 (SPHK1) modulates the proliferation, apoptosis and differentiation of keratinocytes through the regulation of ceramide and sphingosine-1-phosphate levels. However, studies on the expression of SPHK1 in human head and neck squamous cell carcinoma (HNSCC) specimens are lacking. Therefore, the aim of the present work was to evaluate SPHK1 expression in human primary HNSCCs and to correlate the results with clinical and anatomopathological parameters. We investigated the expression of this protein by immunohistochemistry performed in tissue microarrays of HNSCC and in an independent cohort of 37 paraffin-embedded specimens. SPHK1 expression was further validated by real-time PCR performed on laser capture-microdissected tissue samples. The positive rate of SPHK1 protein in the cancerous tissues was significantly higher (74%) than that in the nontumor oral tissues (23%), and malignant tissues showed stronger immunoreactivity for SPHK1 than normal matching samples. These results were confirmed by real-time PCR quantification of SPHK1 mRNA. Interestingly, the positive expression of SPHK1 was associated with shorter patient survival time (Kaplan-Meier survival curves) and with the loss of p21 expression. Taken together, these results demonstrate that SPHK1 is upregulated in HNSCC and provide clues of the role SPHK1 might play in tumor progression.


Sujet(s)
Marqueurs biologiques tumoraux/analyse , Carcinome épidermoïde/enzymologie , Tumeurs de la tête et du cou/enzymologie , Phosphotransferases (Alcohol Group Acceptor)/métabolisme , Technique de Western , Carcinome épidermoïde/génétique , Carcinome épidermoïde/mortalité , Carcinome épidermoïde/anatomopathologie , Évolution de la maladie , Expression des gènes , Régulation de l'expression des gènes tumoraux , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/mortalité , Tumeurs de la tête et du cou/anatomopathologie , Humains , Analyse sur microréseau , Phosphotransferases (Alcohol Group Acceptor)/génétique , Réaction de polymérisation en chaîne , Pronostic , ARN messager/génétique , ARN messager/métabolisme , Sphingolipides/métabolisme , Régulation positive
9.
Exp Mol Pathol ; 88(2): 256-64, 2010 Apr.
Article de Anglais | MEDLINE | ID: mdl-20097195

RÉSUMÉ

It has been recently suggested that p300 cytoplasmic redistribution and degradation are important for controlling the availability and activity of the protein as a transcriptional coactivator. As a step towards determining the functional relevance of p300 intracellular redistribution in mammary cancer, we aimed at studying p300 localization in two different animal models of mammary carcinoma as well as in human primary breast carcinoma samples. Analysis of p300 protein levels showed stronger expression in tumor epithelia than in normal mammary gland. Cytoplasmic localization of p300 was observed in malignant cells. Furthermore, cytoplasmic p300 was found in tumor epithelia whereas nuclear localization was observed in normal mammary glands in both animal models and in non-malignant adjacent areas of human breast cancer specimens. Interestingly, proteasomal inhibition induced p300 redistribution to perinuclear inclusion bodies in tumor but not in normal mammary gland-derived cells. These inclusions were confirmed to be aggresomes by doing immunofluorescence for ubiquitin, vimentin and 20S proteasomal subunit. Taken together, these findings show that both the localization of p300 and the recruitment to aggresomes differ between mammary tumors and normal mammary glands, and suggest that the formation of these inclusions could be a potential target for therapeutic intervention.


Sujet(s)
Tumeurs du sein/métabolisme , Région mammaire/métabolisme , Protéine p300-E1A/métabolisme , Adénocarcinome/génétique , Animaux , Technique de Western , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , Cytoplasme/métabolisme , Cytoplasme/anatomopathologie , Protéine p300-E1A/génétique , Femelle , Humains , Immunohistochimie , Corps d'inclusion/métabolisme , Corps d'inclusion/anatomopathologie , Glandes mammaires animales/métabolisme , Tumeurs mammaires de l'animal/génétique , Souris , Souris transgéniques , Proteasome endopeptidase complex/génétique , Proteasome endopeptidase complex/métabolisme , Valeurs de référence , Transcription génétique , Ubiquitine/métabolisme , Vimentine/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...