Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Viruses ; 15(12)2023 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-38140682

RESUMEN

Syncytin-1 and -2 are glycoproteins encoded by human endogenous retrovirus (hERV) that, through their fusogenic properties, are needed for the formation of the placental syncytiotrophoblast. Previous studies suggested that these proteins, in addition to the EnvP(b) envelope protein, are also involved in other cell fusion events. Since galectin-1 is a ß-galactoside-binding protein associated with cytotrophoblast fusion during placental development, we previously tested its effect on Syncytin-mediated cell fusion and showed that this protein differently modulates the fusogenic potential of Syncytin-1 and -2. Herein, we were interested in comparing the impact of galectin-1 on hERV envelope proteins in different cellular contexts. Using a syncytium assay, we first demonstrated that galectin-1 increased the fusion of Syncytin-2- and EnvP(b)-expressing cells. We then tested the infectivity of Syncytin-1 and -2 vs. VSV-G-pseudotyped viruses toward Cos-7 and various human cell lines. In the presence of galectin-1, infection of Syncytin-2-pseudotyped viruses augmented for all cell lines. In contrast, the impact of galectin-1 on the infectivity of Syncytin-1-pseudotyped viruses varied, being cell- and dose-dependent. In this study, we report the functional associations between three hERV envelope proteins and galectin-1, which should provide information on the fusogenic activity of these proteins in the placenta and other biological and pathological processes.


Asunto(s)
Retrovirus Endógenos , Placenta , Femenino , Humanos , Embarazo , Línea Celular , Retrovirus Endógenos/metabolismo , Galectina 1/metabolismo , Productos del Gen env/genética , Placenta/metabolismo , Trofoblastos/metabolismo , Fusión Celular
2.
Antioxidants (Basel) ; 12(5)2023 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-37237893

RESUMEN

Apolipoprotein D (ApoD) is lipocalin able to bind hydrophobic ligands. The APOD gene is upregulated in a number of pathologies, including Alzheimer's disease, Parkinson's disease, cancer, and hypothyroidism. Upregulation of ApoD is linked to decreased oxidative stress and inflammation in several models, including humans, mice, Drosophila melanogaster and plants. Studies suggest that the mechanism through which ApoD modulates oxidative stress and regulate inflammation is via its capacity to bind arachidonic acid (ARA). This polyunsaturated omega-6 fatty acid can be metabolised to generate large variety of pro-inflammatory mediators. ApoD serves as a sequester, blocking and/or altering arachidonic metabolism. In recent studies of diet-induced obesity, ApoD has been shown to modulate lipid mediators derived from ARA, but also from eicosapentaenoic acid and docosahexaenoic acid in an anti-inflammatory way. High levels of ApoD have also been linked to better metabolic health and inflammatory state in the round ligament of morbidly obese women. Since ApoD expression is upregulated in numerous diseases, it might serve as a therapeutic agent against pathologies aggravated by OS and inflammation such as many obesity comorbidities. This review will present the most recent findings underlying the central role of ApoD in the modulation of both OS and inflammation.

3.
Front Microbiol ; 13: 988944, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36532440

RESUMEN

Human T-cell leukemia virus type 1 is the causative agent of HTLV-1-associated myelopathy/tropical spastic paraparesis and adult T-cell leukemia-lymphoma (ATL). The HTLV-1 basic leucine zipper factor (HBZ) has been associated to the cancer-inducing properties of this virus, although the exact mechanism is unknown. In this study, we identified nucleophosmin (NPM1/B23) as a new interaction partner of HBZ. We show that sHBZ and the less abundant uHBZ isoform interact with nucleolar NPM1/B23 in infected cells and HTLV-1 positive patient cells, unlike equivalent antisense proteins of related non-leukemogenic HTLV-2, -3 and-4 viruses. We further demonstrate that sHBZ association to NPM1/B23 is sensitive to RNase. Interestingly, sHBZ was shown to interact with its own RNA. Through siRNA and overexpression experiments, we further provide evidence that NPM1/B23 acts negatively on viral gene expression with potential impact on cell transformation. Our results hence provide a new insight over HBZ-binding partners in relation to cellular localization and potential function on cell proliferation and should lead to a better understanding of the link between HBZ and ATL development.

4.
Gene ; 756: 144874, 2020 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-32554047

RESUMEN

ApoD is a 25 to 30 kDa glycosylated protein, member of the lipocalin superfamily. As a transporter of several small hydrophobic molecules, its known biological functions are mostly associated to lipid metabolism and neuroprotection. ApoD is a multi-ligand, multi-function protein that is involved lipid trafficking, food intake, inflammation, antioxidative response and development and in different types of cancers. An important aspect of ApoD's role in lipid metabolism appears to involve the transport of arachidonic acid, and the modulation of eicosanoid production and delivery in metabolic tissues. ApoD expression in metabolic tissues has been associated positively and negatively with insulin sensitivity and glucose homeostasis in a tissue dependent manner. ApoD levels rise considerably in association with aging and neuropathologies such as Alzheimer's disease, stroke, meningoencephalitis, moto-neuron disease, multiple sclerosis, schizophrenia and Parkinson's disease. ApoD is also modulated in several animal models of nervous system injury/pathology.


Asunto(s)
Apolipoproteínas D/metabolismo , Animales , Apolipoproteínas D/química , Apolipoproteínas D/genética , Desarrollo Embrionario , Humanos , Neoplasias/metabolismo , Sistema Nervioso/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Especificidad de Órganos
5.
Breast Cancer ; 27(4): 594-606, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31993937

RESUMEN

BACKGROUND: Breast cancer is the most common cancer in women. Despite high survival rates in Western countries, treatments are less effective in metastatic cases and triple-negative breast cancer (TNBC) patient survival is the shortest across breast cancer subtypes. High expression levels of stearoyl-CoA desaturase-1 (SCD1) have been reported in breast cancer. The SCD1 enzyme catalyzes the formation of oleic acid (OA), a lipid stimulating the migration of metastatic breast cancer cells. Phospholipase activity is also implicated in breast cancer metastasis, notably phospholipase D (PLD). METHODS: Kaplan-Meier survival plots generated from gene expression databases were used to analyze the involvement of SCD1 and PLD in several cancer subtypes. SCD1 enzymatic activity was modulated with a pharmaceutical inhibitor or by OA treatment (to mimic SCD1 over-activity) in three breast cancer cell lines: TNBC-derived MDA-MB-231 cells as well as non-TNBC MCF-7 and T47D cells. Cell morphology and migration properties were characterized by various complementary methods. RESULTS: Our survival analyses suggest that SCD1 and PLD2 expression in the primary tumor are both associated to metastasis-related morbid outcomes in breast cancer patients. We show that modulation of SCD1 activity is associated with the modification of TNBC cell migration properties, including changes in speed, direction and cell morphology. Cell migration properties are regulated by SCD1 activity through a PLD-mTOR/p70S6K signaling pathway. These effects are not observed in non-TNBC cell lines. CONCLUSION: Our results establish a key role for the lipid desaturase SCD1 and delineate an OA-PLD-mTOR/p70S6K signaling pathway in TNBC-derived MDA-MB-231 cell migration.


Asunto(s)
Movimiento Celular , Estearoil-CoA Desaturasa/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Línea Celular Tumoral , Conjuntos de Datos como Asunto , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Metástasis de la Neoplasia , Ácido Oléico/metabolismo , Fosfolipasa D/antagonistas & inhibidores , Fosfolipasa D/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Neoplasias de la Mama Triple Negativas/mortalidad , Neoplasias de la Mama Triple Negativas/cirugía
6.
J Virol ; 93(2)2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30404795

RESUMEN

The existence of the antisense transcript-encoded HIV-1 antisense protein (ASP) was recently reinforced by in silico analyses providing evidence for recent appearance of this gene in the viral genome. Our previous studies led to the detection of ASP in various cell lines by Western blotting, flow cytometry, and confocal microscopy analyses and reported that it induced autophagy, potentially through multimer formation. Here, our goals were to assess autophagy induction by ASP from different clades and to identify the implicated autophagy factors. We first demonstrated that ASP formed multimers, partly through its amino-terminal region and cysteine residues. Removal of this region was further associated with lower induction of autophagy, as assessed by autophagosome formation. ASPs from different clades (A, B, C, D, and G) were tested next and were detected in monomeric and multimeric forms at various levels, and all induced autophagy (clade A ASP was less efficient), as determined by LC3-II and p62 (SQSTM1) levels. Furthermore, CRISPR-based knockout of ATG5, ATG7, and p62 genes led to increased ASP levels. Confocal microscopy analyses showed that ASP colocalized with p62 and LC3-II in autophagosome-like structures. Coimmunoprecipitation experiments further demonstrated that p62 associated with ASP through its PB1 domain. Interestingly, immunoprecipitation experiments supported the idea that ASP is ubiquitinated and that ubiquitination was modulating its stability. We are thus suggesting that ASP induces autophagy through p62 interaction and that its abundance is controlled by autophagy, in which ubiquitin plays an important role. Understanding the mechanisms underlying ASP degradation is essential to better assess its function.IMPORTANCE In the present study, we provide the first evidence that a new HIV-1 protein termed ASP derived from different clades acts similarly in inducing autophagy, an important cellular process implicated in the degradation of excess or defective cellular material. We have gained further knowledge on the mechanism mediating the activation of autophagy. Our studies have important ramifications in the understanding of viral replication and the pathogenesis associated with HIV-1 in infected individuals. Indeed, autophagy is implicated in antigen presentation during immune response and could thus be rendered inefficient in infected cells, such as dendritic cells. Furthermore, a possible link with HIV-1-associated neurological disorder (HAND) might also be a possible association with the capacity of ASP to induce autophagy. Our studies hence demonstrate the importance in conducting further studies on this protein as it could represent a new interesting target for antiretroviral therapies and vaccine design.


Asunto(s)
VIH-1/metabolismo , Proteína Sequestosoma-1/química , Proteína Sequestosoma-1/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Animales , Autofagia , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Modelos Moleculares , Dominios Proteicos , Multimerización de Proteína , Ubiquitinación
7.
Free Radic Biol Med ; 130: 499-511, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30445127

RESUMEN

Oxidative stress plays a pivotal and early role in the pathophysiology of Alzheimer's disease (AD). There is convincing evidence that oxidative alterations in AD and in mild cognitive impairment (MCI) patients are not limited to the brain but are extended to the blood compartment. However, the oxidative pattern in plasma is still inconclusive. Moreover, their potential association with the clinical scores MMSE (Mini-Mental State Examination) and MoCA (Montreal Cognitive Assessment) is poorly investigated. The aim of our study was to establish a pattern of blood-based redox alterations in prodromal AD and their evolution during the progression of the disease. Our results showed a reduction in the total antioxidant capacity (TAC) and an increase of the stress-response proteins apolipoprotein J (ApoJ) and Klotho in MCI subjects. For the first time, we evidenced circulating-proteasome activity. We found that the alteration of the circulating-proteasome activity is associated with the accumulation of oxidized proteins in plasma form early AD. Interestingly, the TAC, the levels of vitamin D and the activity of proteasome were positively associated to the clinical scores MMSE and MoCA. The levels of protein carbonyls and of ApoJ were negatively associated to the MMSE and MoCA scores. The levels of apolipoprotein D (ApoD) were not different between groups. Interestingly, the receiver operating characteristic (ROC) curves analysis indicated that these redox markers provide a fair classification of different groups with high accuracy. Overall, our results strengthen the notion that some specific oxidative markers could be considered as non-invasive blood-based biomarkers for an early MCI diagnosis and AD progression.


Asunto(s)
Enfermedad de Alzheimer/sangre , Biomarcadores/sangre , Encéfalo/metabolismo , Disfunción Cognitiva/sangre , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Apolipoproteínas D/sangre , Encéfalo/patología , Clusterina/sangre , Disfunción Cognitiva/patología , Progresión de la Enfermedad , Femenino , Humanos , Proteínas Klotho , Masculino , Proteínas de la Membrana/sangre , Oxidación-Reducción , Estrés Oxidativo/genética , Complejo de la Endopetidasa Proteasomal/genética , Vitamina D/sangre
8.
Endocrine ; 61(2): 248-257, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29869155

RESUMEN

PURPOSE: Apolipoprotein D (ApoD) is a lipocalin participating in lipid transport. It binds to a variety of ligands, with a higher affinity for arachidonic acid, and is thought to have a diverse array of functions. We investigated a potential role for ApoD in insulin sensitivity, inflammation, and thrombosis-processes related to lipid metabolism-in severely obese women. METHODS: We measured ApoD expression in a cohort of 44 severely obese women including dysmetabolic and non-dysmetabolic patients. Physical and metabolic characteristics of these women were determined from anthropometric measurements and blood samples. ApoD was quantified at the mRNA and protein levels in samples from three intra-abdominal adipose tissues (AT): omental, mesenteric and round ligament (RL). RESULTS: ApoD protein levels were highly variable between AT of the same individual. High ApoD protein levels, particularly in the RL depot, were linked to lower plasma insulin levels (-40%, p = 0.015) and insulin resistance (-47%, p = 0.022), and increased insulin sensitivity (+10%, p = 0.008). Lower circulating pro-inflammatory PAI-1 (-39%, p = 0.001), and TNF-α (-19%, p = 0.030) levels were also correlated to high ApoD protein in the RL AT. CONCLUSIONS: ApoD variability between AT was consistent with different accumulation efficiencies and/or metabolic functions according to the anatomic location of fat depots. Most statistically significant correlations implicated ApoD protein levels, in agreement with protein accumulation in target tissues. These correlations associated higher ApoD levels in fat depots with improved metabolic health in severely obese women.


Asunto(s)
Apolipoproteínas D/genética , Inflamación/sangre , Grasa Intraabdominal/metabolismo , Obesidad Mórbida/genética , Obesidad Mórbida/metabolismo , Ligamentos Redondos/metabolismo , Adulto , Apolipoproteínas D/metabolismo , Femenino , Humanos , Inflamación/complicaciones , Inflamación/metabolismo , Mediadores de Inflamación/sangre , Resistencia a la Insulina/genética , Interleucina-6/sangre , Metabolismo de los Lípidos/fisiología , Persona de Mediana Edad , Obesidad Mórbida/complicaciones , Obesidad Mórbida/patología , Inhibidor 1 de Activador Plasminogénico/sangre , Factor de Necrosis Tumoral alfa/sangre , Adulto Joven
9.
Mol Neurobiol ; 54(6): 3948-3963, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-27271124

RESUMEN

Excitotoxicity due to the excessive activation of glutamatergic receptors leads to neuronal dysfunction and death. Excitotoxicity has been implicated in the pathogenesis of a myriad of neurodegenerative diseases with distinct etiologies such as Alzheimer's and Parkinson's. Numerous studies link apolipoprotein D (apoD), a secreted glycoprotein highly expressed in the central nervous system (CNS), to maintain and protect neurons in various mouse models of acute stress and neurodegeneration. Here, we used a mouse model overexpressing human apoD in neurons (H-apoD Tg) to test the neuroprotective effects of apoD in the kainic acid (KA)-lesioned hippocampus. Our results show that apoD overexpression in H-apoD Tg mice induces an increased resistance to KA-induced seizures, significantly attenuates inflammatory responses and confers protection against KA-induced cell apoptosis in the hippocampus. The apoD-mediated protection against KA-induced toxicity is imputable in part to increased plasma membrane Ca2+ ATPase type 2 expression (1.7-fold), decreased N-methyl-D-aspartate receptor (NMDAR) subunit NR2B levels (30 %) and lipid metabolism alterations. Indeed, we demonstrate that apoD can attenuate intracellular cholesterol content in primary hippocampal neurons and in brain of H-apoD Tg mice. In addition, apoD can be internalised by neurons and this internalisation is accentuated in ageing and injury conditions. Our results provide additional mechanistic information on the apoD-mediated neuroprotection in neurodegenerative conditions.


Asunto(s)
Apolipoproteínas D/metabolismo , Ácido Kaínico/toxicidad , Neuroprotección , Neurotoxinas/toxicidad , Animales , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Colesterol/metabolismo , Endocitosis , Hipocampo/efectos de los fármacos , Hipocampo/patología , Humanos , Inflamación/patología , Ratones Transgénicos , Modelos Biológicos , Neuronas/metabolismo , Neuroprotección/efectos de los fármacos , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Subunidades de Proteína/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsiones/patología , Regulación hacia Arriba/efectos de los fármacos
10.
Metabolism ; 65(9): 1247-58, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27506732

RESUMEN

BACKGROUND: Apolipoprotein D (ApoD) is a member of the lipocalin family known to transport small hydrophobic ligands. A major site of ApoD expression in mice is the central nervous system where evidence suggests that it plays a protective role. Gene expression of ApoD was reported in bone-forming osteoblasts but its impact on bone metabolism remains undocumented. METHODS: We compared basic bone parameters of ApoD(-/-) (null) and transgenic (tg) mice to wild-type (wt) littermates through microCT and histochemistry, as well as ApoD expression and secretion in osteoblasts under various culture conditions through real-time PCR and immunoblotting. RESULTS: ApoD-null females displayed progressive bone loss with aging, resulting in a 50% reduction in trabecular bone volume and a 23% reduction in cortical bone volume by 9months of age. Only cortical bone volume was significantly reduced in ApoD-null males by an average of 24%. Histochemistry indicated significantly higher osteoblast surface and number of osteoclasts in femora from ApoD-null females. ApoD gene expression was confirmed in primary cultures of bone marrow mesenchymal cells (MSC), with higher expression levels in MSC from females compared to males. ApoD-null MSC exhibited impaired proliferation and differentiation potentials. Moreover, exogenous ApoD partially rescued the osteogenic potential of null MSC, which were shown to readily uptake the protein from media. ApoD expression was upregulated under low proliferation conditions, by contact inhibition and osteoblastic differentiation in MC3T3-E1 osteoblast-like cells. CONCLUSION: Our results indicate that ApoD influences bone metabolism in mice in a gender-specific manner, potentially through an auto-/paracrine pathway.


Asunto(s)
Envejecimiento/genética , Apolipoproteínas D/deficiencia , Desarrollo Óseo/genética , Remodelación Ósea/genética , Osteoblastos , Células 3T3 , Animales , Apolipoproteínas D/genética , Apolipoproteínas D/metabolismo , Células de la Médula Ósea/metabolismo , Ciclo Celular/genética , Diferenciación Celular , Proliferación Celular , Femenino , Fémur/citología , Fémur/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Osteoblastos/metabolismo , Cultivo Primario de Células
11.
PLoS One ; 10(6): e0130230, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26083030

RESUMEN

Transgenic mice (Tg) overexpressing human apolipoprotein D (H-apoD) in the brain are resistant to neurodegeneration. Despite the use of a neuron-specific promoter to generate the Tg mice, they expressed significant levels of H-apoD in both plasma and liver and they slowly develop hepatic steatosis and insulin resistance. We show here that hepatic PPARγ expression in Tg mice is increased by 2-fold compared to wild type (WT) mice. Consequently, PPARγ target genes Plin2 and Cide A/C are overexpressed, leading to increased lipid droplets formation. Expression of the fatty acid transporter CD36, another PPARgamma target, is also increased in Tg mice associated with elevated fatty acid uptake as measured in primary hepatocytes. Elevated expression of AMPK in the liver of Tg leads to phosphorylation of acetyl CoA carboxylase, indicating a decreased activity of the enzyme. Fatty acid synthase expression is also induced but the hepatic lipogenesis measured in vivo is not significantly different between WT and Tg mice. In addition, expression of carnitine palmitoyl transferase 1, the rate-limiting enzyme of beta-oxidation, is slightly upregulated. Finally, we show that overexpressing H-apoD in HepG2 cells in presence of arachidonic acid (AA), the main apoD ligand, increases the transcriptional activity of PPARγ. Supporting the role of apoD in AA transport, we observed enrichment in hepatic AA and a decrease in plasmatic AA concentration. Taken together, our results demonstrate that the hepatic steatosis observed in apoD Tg mice is a consequence of increased PPARγ transcriptional activity by AA leading to increased fatty acid uptake by the liver.


Asunto(s)
Apolipoproteínas D/genética , Ácidos Grasos/metabolismo , Hígado Graso/genética , Hígado Graso/metabolismo , PPAR gamma/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Ácido Araquidónico/sangre , Ácido Araquidónico/metabolismo , Ácido Araquidónico/farmacología , Proteínas Potenciadoras de Unión a CCAAT/genética , Antígenos CD36/metabolismo , Ácido Graso Sintasas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/metabolismo , Lipogénesis/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , PPAR gamma/genética , Perilipina-2 , Transporte de Proteínas/efectos de los fármacos , Proteínas/metabolismo
12.
J Biol Chem ; 290(26): 16077-87, 2015 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-25918162

RESUMEN

Apolipoprotein D (apoD), a member of the lipocalin family, is a 29-kDa secreted glycoprotein that binds and transports small lipophilic molecules. Expressed in several tissues, apoD is up-regulated under different stress stimuli and in a variety of pathologies. Numerous studies have revealed that overexpression of apoD led to neuroprotection in various mouse models of acute stress and neurodegeneration. This multifunctional protein is internalized in several cells types, but the specific internalization mechanism remains unknown. In this study, we demonstrate that the internalization of apoD involves a specific cell surface receptor in 293T cells, identified as the transmembrane glycoprotein basigin (BSG, CD147); more particularly, its low glycosylated form. Our results show that internalized apoD colocalizes with BSG into vesicular compartments. Down-regulation of BSG disrupted the internalization of apoD in cells. In contrast, overexpression of basigin in SH-5YSY cells, which poorly express BSG, restored the uptake of apoD. Cyclophilin A, a known ligand of BSG, competitively reduced apoD internalization, confirming that BSG is a key player in the apoD internalization process. In summary, our results demonstrate that basigin is very likely the apoD receptor and provide additional clues on the mechanisms involved in apoD-mediated functions, including neuroprotection.


Asunto(s)
Apolipoproteínas D/metabolismo , Basigina/metabolismo , Apolipoproteínas D/genética , Basigina/genética , Línea Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Humanos , Transporte de Proteínas
13.
PLoS One ; 10(3): e0121468, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25781974

RESUMEN

Syncytin-2 is encoded by the envelope gene of Endogenous Retrovirus-FRD (ERVFRD-1) and plays a critical role in fusion of placental trophoblasts leading to the formation of the multinucleated syncytiotrophoblast. Its expression is consequently regulated in a strict manner. In the present study, we have identified a forskolin-responsive region located between positions -300 to -150 in the Syncytin-2 promoter region. This 150 bp region in the context of a minimal promoter mediated an 80-fold induction of promoter activity following forskolin stimulation. EMSA analyses with competition experiments with nuclear extracts from forskolin-stimulated BeWo cells demonstrated that the -211 to -177 region specifically bound two forskolin-induced complexes, one of them containing a CRE/AP-1-like motif. Site-directed mutagenesis of the CRE/AP-1 binding site in the context of the Syncytin-2 promoter or a heterologous promoter showed that this motif was mostly essential for forskolin-induced promoter activity. Transfection experiments with dominant negative mutants and constitutively activated CREB expression vectors in addition to Chromatin Immunoprecipitation suggested that a CREB family member, CREB2 was binding and acting through the CRE/AP-1 motif. We further demonstrated the binding of JunD to this same motif. Similar to forskolin and soluble cAMP, CREB2 and JunD overexpression induced Syncytin-2 promoter activity in a CRE/AP-1-dependent manner and Syncytin-2 expression. In addition, BeWo cell fusion was induced by both CREB2 and JunD overexpression, while being repressed following silencing of either gene. These results thereby demonstrate that induced expression of Syncytin-2 is highly dependent on the interaction of bZIP-containing transcription factors to a CRE/AP-1 motif and that this element is important for the regulation of Syncytin-2 expression, which results in the formation of the peripheral syncytiotrophoblast layer.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Modelos Biológicos , Motivos de Nucleótidos/fisiología , Proteínas Gestacionales/biosíntesis , Elementos de Respuesta/fisiología , Trofoblastos/metabolismo , Adulto , Fusión Celular , Línea Celular Tumoral , Femenino , Humanos , Proteínas Gestacionales/genética , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Trofoblastos/citología
14.
Cell Oncol (Dordr) ; 37(3): 179-91, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24916915

RESUMEN

BACKGROUND: Previously, we found that the Graffi murine leukemia virus (MuLV) is able to induce a wide spectrum of hematologic malignancies in vivo. Using high-density oligonucleotide microarrays, we established the gene expression profiles of several of these malignancies, thereby specifically focusing on genes deregulated in the lymphoid sub-types. We observed over-expression of a variety of genes, including Arntl2, Bfsp2, Gfra2, Gpm6a, Gpm6b, Nln, Fbln1, Bmp7, Etv5 and Celsr1 and, in addition, provided evidence that Fmn2 and Parm-1 may act as novel oncogenes. In the present study, we assessed the expression patterns of eight selected human homologs of these genes in primary human B-cell malignancies, and explored the putative oncogenic potential of GPM6A and GPM6B. METHODS: The gene expression levels of the selected human homologs were tested in human B-cell malignancies by semi-quantitative RT-PCR. The protein expression profiles of human GPM6A and GPM6B were analyzed by Western blotting. The localization and the effect of GPM6A and GPM6B on the cytoskeleton were determined using confocal and indirect immunofluorescence microscopy. To confirm the oncogenic potential of GPM6A and GPM6B, classical colony formation assays in soft agar and focus forming assays were used. The effects of these proteins on the cell cycle were assessed by flow cytometry analysis. RESULTS: Using semi-quantitative RT-PCR, we found that most of the primary B-cell malignancies assessed showed altered expression patterns of the genes tested, including GPM6A and GPM6B. Using confocal microscopy, we found that the GPM6A protein (isoform 3) exhibits a punctate cytoplasmic localization and that the GPM6B protein (isoform 4) exhibits a peri-nuclear and punctate cytoplasmic localization. Interestingly, we found that exogenous over-expression of both proteins in NIH/3T3 cells alters the actin and microtubule networks and induces the formation of long filopodia-like protrusions. Additionally, we found that these over-expressing NIH/3T3 cells exhibit anchorage-independent growth and enhanced proliferation rates. Cellular transformation (i.e., loss of contact inhibition) was, however, only observed after exogenous over-expression of GPM6B. CONCLUSIONS: Our results indicate that several human homologs of the genes found to be deregulated in Graffi MuLV experimental mouse models may serve as candidate biomarkers for human B-cell malignancies. In addition, we found that GPM6A and GPM6B may act as novel oncogenes in the development of these malignancies.


Asunto(s)
Glicoproteínas de Membrana/genética , Proteínas del Tejido Nervioso/genética , Proteínas Oncogénicas/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Animales , Western Blotting , Adhesión Celular/genética , Ciclo Celular/genética , Proliferación Celular , Niño , Citoesqueleto/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Microscopía Confocal , Microscopía Fluorescente , Células 3T3 NIH , Proteínas del Tejido Nervioso/metabolismo , Proteínas Oncogénicas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
15.
Int J Oncol ; 45(2): 485-92, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24827738

RESUMEN

In normal tissues, energy-providing lipids come principally from circulating lipids. However, in growing tumors, energy supply is mainly provided by lipids coming from de novo synthesis. It is not surprising to see elevated expression of several lipogenic genes in tumors from different origins. The role of lipogenic genes in the establishment of the primary tumor has been clearly established. A large number of studies demonstrate a role of fatty acid synthase in the activation of cell cycle and inhibition of apoptosis in tumor cells. Other lipogenic genes such as the acetyl CoA carboxylase (ACC) and the stearoyl CoA desaturase 1 (SCD1) are highly expressed in primary tumors and also appear to play a role in their development. However, the role of lipogenesis in the metastatic process is less clear. In the present review, we aim to present the most recent evidences for the key role of lipogenic enzymes in the metastatic process and in epithelial to mesenchymal transition.


Asunto(s)
Lipogénesis/fisiología , Neoplasias/metabolismo , Animales , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal/fisiología , Humanos , Neoplasias/patología , Transducción de Señal/fisiología
16.
Mol Cancer ; 12: 84, 2013 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-23902727

RESUMEN

BACKGROUND: The Graffi murine retrovirus is a powerful tool to find leukemia associated oncogenes. Using DNA microarrays, we recently identified several genes specifically deregulated in T- and B-leukemias induced by this virus. RESULTS: In the present study, probsets associated with T-CD8+ leukemias were analyzed and we validated the expression profile of the Parm-1 gene. PARM-1 is a member of the mucin family. We showed that human PARM-1 is an intact secreted protein accumulating predominantly, such as murine PARM-1, at the Golgi and in the early and late endosomes. PARM-1 colocalization with α-tubulin suggests that its trafficking within the cell involves the microtubule cytoskeleton. Also, the protein co-localizes with caveolin-1 which probably mediates its internalization. Transient transfection of both mouse and human Parm-1 cDNAs conferred anchorage- and serum-independent growth and enhanced cell proliferation. Moreover, deletion mutants of human PARM-1 without either extracellular or cytoplasmic portions seem to retain the ability to induce anchorage-independent growth of NIH/3T3 cells. In addition, PARM-1 increases ERK1/2, but more importantly AKT and STAT3 phosphorylation. CONCLUSIONS: Our results strongly suggest the oncogenic potential of PARM-1.


Asunto(s)
Proteína de Unión a Andrógenos/genética , Oncogenes , Proteína de Unión a Andrógenos/metabolismo , Animales , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Caveolina 1 , Proliferación Celular , Transformación Celular Neoplásica/genética , Endosomas/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Aparato de Golgi/metabolismo , Humanos , Leucemia de Células T/genética , Leucemia de Células T/metabolismo , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Mutación , Células 3T3 NIH , Unión Proteica , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Tubulina (Proteína)/metabolismo
17.
Mol Neurobiol ; 48(3): 669-80, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23715769

RESUMEN

Apolipoprotein D (ApoD) gene expression is increased in several neurological disorders such as Alzheimer's disease (AD) and multiple sclerosis. We previously showed that transgenic mice that overexpress human ApoD show a better resistance against paraquat or OC43 coronavirus-induced neurodegeneration. Here, we identified several nuclear factors from the cortex of control and OC43-infected mice which bind a fragment of the proximal ApoD promoter in vitro. Of interest, we detected apolipoprotein E (ApoE). Human ApoE consists of three isoforms (E2, E3, and E4) with the E4 and E2 alleles representing a greater and a lower risk for developping AD, respectively. Our results show that ApoE is located in the nucleus and on the ApoD promoter in human hepatic and glioblastoma cells lines. Furthermore, overexpression of ApoE3 and ApoE4 isoforms but not ApoE2 significantly inhibited the ApoD promoter activity in U87 cells (E3/E3 genotype) cultured under normal or different stress conditions while ApoE knock-down by siRNA had a converse effect. Consistent with these results, we also demonstrated by ChIP assay that E3 and E4 isoforms, but not E2, bind the ApoD promoter. Moreover, using the Allen Brain Atlas in situ hybridization database, we observed an inverse correlation between ApoD and ApoE mRNA expression during development and in several regions of the mouse brain, notably in the cortex, hippocampus, plexus choroid, and cerebellum. This negative correlation was also observed for cortex layers IV-VI based on a new Transcriptomic Atlas of the Mouse Neocortical Layers. These findings reveal a new function for ApoE by regulating ApoD gene expression.


Asunto(s)
Apolipoproteína E3/metabolismo , Apolipoproteína E4/metabolismo , Apolipoproteínas D/genética , Regiones Promotoras Genéticas , Animales , Secuencia de Bases , Línea Celular Tumoral , Núcleo Celular/metabolismo , Corteza Cerebral/metabolismo , Regulación de la Expresión Génica , Humanos , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteínas Nucleares/aislamiento & purificación , Proteínas Nucleares/metabolismo , Unión Proteica , Isoformas de Proteínas/metabolismo , Transporte de Proteínas
18.
Cancer Sci ; 104(1): 36-42, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23013158

RESUMEN

Stearoyl-CoA desaturase-1 (SCD1) is an endoplasmic reticulum anchored enzyme catalyzing the synthesis of monounsaturated fatty acids, mainly palmytoleyl-CoA and oleyl-CoA. Recent studies have revealed a function for SCD1 in the modulation of signaling processes related to cell proliferation, survival and transformation to cancer. We used MCF7 and MDA-MB-231 cells to analyze the role of SCD1 in the metastatic acquisition of breast cancer cells. Silencing SCD1 expression in breast cancer cells has no effect on cell viability but the levels of cell proliferation, cell cycle genes' expressions and the phosphorylation state of ERK1/2 MAPK are significantly reduced. Decreasing SCD1 expression also reduces the level of GSK3 phosphorylation, indicating higher activity of the kinase. Using cells fractionation, immunofluorescence and a ß-catenin/TCF-responsive reporter construct, we demonstrate that lowering SCD1 expression leads to a decrease of ß-catenin amounts within the nucleus and to inhibition of its transactivation capacity. Moreover, MDA-MB-231 cells transfected with the SCD1 siRNA show a lower invasive potential than the control cells. Taken together, our data demonstrate that low SCD1 expression is associated with a decrease in the proliferation rate of breast cancer cells associated with a decrease in ERK1/2 activation. SCD1 silencing also inhibits GSK3 phosphorylation, lowering ß-catenin translocation to the nucleus, and, subsequently, its transactivation capacity and the expression of its target genes. Finally, we show that silencing SCD1 impairs the epithelial to mesenchymal transition-like behavior of the cells, a characteristic of metastatic breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Invasividad Neoplásica , Transducción de Señal , Estearoil-CoA Desaturasa/metabolismo , beta Catenina/metabolismo , Neoplasias de la Mama/patología , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Transformación Celular Neoplásica , Retículo Endoplásmico/enzimología , Transición Epitelial-Mesenquimal/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ácidos Grasos Monoinsaturados/metabolismo , Femenino , Glucógeno Sintasa Quinasas/metabolismo , Humanos , Células MCF-7 , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño , Estearoil-CoA Desaturasa/genética
19.
Reprod Sci ; 19(4): 374-82, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22344723

RESUMEN

The syncytiotrophoblast is a multinuclear cell layer maintained through fusion events with cytotrophoblasts and plays a key role in the properties of the placenta. Monitoring fusion in this cell layer is important in studies aimed at understanding its function. We herein propose a new fusion assay based on the transactivating potential of the human immunodeficiency virus type 1 (HIV-1) Tat protein on its promoter present in the long terminal repeat (LTR) region. We used 2 BeWo cell populations, one stably transfected with the HIV-1 LTR positioned upstream of the luciferase gene and the other stably transfected with a Tat expression vector. Both stable cell lines were responsive to Tat-mediated LTR transactivation and demonstrated normal fusion and human chorionic gonadotropin (hCG) secretion upon stimulation. When both BeWo cell lines were cocultured, forskolin-mediated induction of fusion led to an increase in luciferase activity, which was sensitive to anti-syncytin 1 and -2 antibodies and syncytin 2 small interfering RNAs (siRNAs). Similar results were obtained in primary trophoblasts. Our results highlight the effectiveness and accuracy of this new quantification assay for trophoblast fusion.


Asunto(s)
Luciferasas/metabolismo , Placenta/fisiología , Trofoblastos/fisiología , Línea Celular Tumoral , Gonadotropina Coriónica/metabolismo , Técnicas de Cocultivo , Colforsina/farmacología , Femenino , Duplicado del Terminal Largo de VIH , Humanos , Luciferasas/genética , Microscopía Confocal , Placenta/citología , Embarazo , Regiones Promotoras Genéticas , Transfección , Trofoblastos/citología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
20.
Viruses ; 3(11): 2146-59, 2011 11.
Artículo en Inglés | MEDLINE | ID: mdl-22163338

RESUMEN

Human endogenous retroviruses (HERVs) represent approximately 8% of our genome. HERVs influence cellular gene expression and contribute to normal physiological processes such as cellular differentiation and morphogenesis. HERVs have also been associated with certain pathological conditions, including cancer and neurodegenerative diseases. As HTLV-1 causes adult T-cell leukemia and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) and has been shown to modulate host gene expression mainly through the expression of the powerful Tax transactivator, herein we were interested in looking at the potential modulation capacity of HTLV-1 Tax on HERV expression. In order to evaluate the promoter activity of different HERV LTRs, pHERV-LTR-luc constructs were co-transfected in Jurkat T-cells with a Tax expression vector. Tax expression potently increased the LTR activity of HERV-W8 and HERV-H (MC16). In parallel, Jurkat cells were also stimulated with different T-cell-activating agents and HERV LTRs were observed to respond to different combination of Forskolin, bpV[pic] a protein tyrosine phosphatase inhibitor, and PMA. Transfection of expression vectors for different Tax mutants in Jurkat cells showed that several transcription factors including CREB appeared to be important for HERV-W8 LTR activation. Deletion mutants were derived from the HERV-W8 LTR and the region from -137 to -123 was found to be important for LTR response following Tax expression in Jurkat cells, while a different region was shown to be required in cells treated with activators. Our results thus demonstrated that HTLV-1 Tax activates several HERV LTRs. This raises the possibility that upregulated HERV expression could be involved in diseases associated with HTLV-1 infection.


Asunto(s)
Retrovirus Endógenos/genética , Productos del Gen tax/metabolismo , Duplicado del Terminal Largo de VIH , Infecciones por HTLV-I/virología , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Activación de Linfocitos , Linfocitos T/inmunología , Activación Transcripcional , Línea Celular , Retrovirus Endógenos/metabolismo , Regulación Viral de la Expresión Génica , Productos del Gen tax/genética , Infecciones por HTLV-I/inmunología , Virus Linfotrópico T Tipo 1 Humano/genética , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA