Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Redox Biol ; 71: 103074, 2024 May.
Article in English | MEDLINE | ID: mdl-38367511

ABSTRACT

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.


Subject(s)
Ferroptosis , Iron Overload , Motor Disorders , Mice , Animals , Lipid Metabolism , Motor Disorders/metabolism , Iron/metabolism , Lipid Peroxidation , Dopaminergic Neurons/metabolism , Cholesterol/metabolism , Lipids
2.
J Steroid Biochem Mol Biol ; 154: 285-93, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26365558

ABSTRACT

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.


Subject(s)
Breast Neoplasms/pathology , Calcitriol/pharmacology , Neoplasm Metastasis/prevention & control , Organophosphonates/pharmacology , Animals , Calcitriol/analogs & derivatives , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C
3.
Exp Mol Pathol ; 97(3): 411-24, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25240203

ABSTRACT

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.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , E1A-Associated p300 Protein/metabolism , Animals , Apoptosis/physiology , Blotting, Western , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Movement , Cell Proliferation/physiology , Cytoplasm/chemistry , Cytoplasm/metabolism , Disease Models, Animal , Female , Fluorescent Antibody Technique , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Microscopy, Confocal
4.
J Neurooncol ; 118(1): 49-60, 2014 May.
Article in English | MEDLINE | ID: mdl-24584679

ABSTRACT

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.


Subject(s)
Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/genetics , Glioblastoma/metabolism , Receptors, Calcitriol/metabolism , Adult , Age Factors , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Calcitriol/pharmacology , Calcium Channel Agonists/pharmacology , Cell Line, Tumor , Cell Movement/genetics , Cell Survival/drug effects , Cell Survival/physiology , Cyclin D1/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Male , Middle Aged , Oncogene Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sex Factors , Time Factors , Tissue Array Analysis
5.
Tumour Biol ; 35(3): 2803-15, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24234335

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/analysis , Brain Neoplasms/enzymology , Glioma/enzymology , Heme Oxygenase-1/biosynthesis , Astrocytoma/enzymology , Astrocytoma/mortality , Astrocytoma/pathology , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Fluorescent Antibody Technique , Glioma/mortality , Glioma/pathology , Heme Oxygenase-1/analysis , Humans , Immunoblotting , Immunohistochemistry , Kaplan-Meier Estimate , Prognosis , Real-Time Polymerase Chain Reaction , Tissue Array Analysis
7.
Exp Mol Pathol ; 93(2): 237-45, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22580187

ABSTRACT

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.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Head and Neck Neoplasms/enzymology , Heme Oxygenase-1/metabolism , Aged , Animals , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/genetics , Disease Models, Animal , Disease Progression , Female , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/genetics , Heme Oxygenase-1/genetics , Humans , Male , Mice , Middle Aged , Paraffin Embedding , Polymerase Chain Reaction , Prognosis , RNA, Messenger/metabolism , Tissue Array Analysis
8.
Cells Tissues Organs ; 192(5): 314-24, 2010.
Article in English | MEDLINE | ID: mdl-20606403

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/enzymology , Head and Neck Neoplasms/enzymology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Disease Progression , Gene Expression , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/pathology , Humans , Microarray Analysis , Phosphotransferases (Alcohol Group Acceptor)/genetics , Polymerase Chain Reaction , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sphingolipids/metabolism , Up-Regulation
9.
Exp Mol Pathol ; 88(2): 256-64, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20097195

ABSTRACT

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.


Subject(s)
Breast Neoplasms/metabolism , Breast/metabolism , E1A-Associated p300 Protein/metabolism , Adenocarcinoma/genetics , Animals , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cytoplasm/metabolism , Cytoplasm/pathology , E1A-Associated p300 Protein/genetics , Female , Humans , Immunohistochemistry , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/genetics , Mice , Mice, Transgenic , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Reference Values , Transcription, Genetic , Ubiquitin/metabolism , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...