Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 47
Filtrer
1.
Int J Mol Sci ; 22(19)2021 Sep 29.
Article de Anglais | MEDLINE | ID: mdl-34638901

RÉSUMÉ

Among the mechanisms leading to progression to Adult T-cell Leukaemia/Lymphoma in Human T-cell Leukaemia Virus type 1 (HTLV-1)-infected subjects, the contribution of stromal components remains poorly understood. To dissect the role of fibroblasts in HTLV-1-mediated lymphomagenesis, transcriptome studies, cytofluorimetric and qRT-PCR analyses of surface and intracellular markers linked to plasticity and stemness in coculture, and in vivo experiments were performed. A transcriptomic comparison between a more lymphomagenic (C91/III) and the parental (C91/PL) cell line evidenced hyperactivation of the PI3K/Akt pathway, confirmed by phospho-ELISA and 2-DE and WB analyses. C91/III cells also showed higher expression of mesenchymal and stemness genes. Short-term coculture with human foreskin fibroblasts (HFF) induced these features in C91/PL cells, and significantly increased not only the cancer stem cells (CSCs)-supporting CD10+GPR77+ HFF subpopulation, but also the percentage of ALDH1bright C91/PL cells. A non-cytotoxic acetylsalicylic acid treatment decreased HFF-induced ALDH1bright C91/PL cells, downregulated mesenchymal and stemness genes in cocultured cells, and delayed lymphoma growth in immunosuppressed mice, thus hindering the supportive activity of HFF on CSCs. These data suggest that crosstalk with HFF significantly intensifies the aggressiveness and plasticity of C91/PL cells, leading to the enrichment in lymphoma-initiating cells. Additional research is needed to better characterize these preliminary findings.


Sujet(s)
Fibroblastes/métabolisme , Analyse de profil d'expression de gènes/méthodes , Régulation de l'expression des gènes tumoraux/génétique , Lymphomes/génétique , Cellules souches tumorales/métabolisme , Animaux , Anti-inflammatoires non stéroïdiens/pharmacologie , Acide acétylsalicylique/pharmacologie , Lignée cellulaire , Cellules cultivées , Techniques de coculture , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/virologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Virus T-lymphotrope humain de type 1/physiologie , Humains , Cellules Jurkat , Lymphomes/traitement médicamenteux , Lymphomes/virologie , Souris de lignée NOD , Souris knockout , Souris SCID , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/virologie , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/génétique , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes
2.
Front Microbiol ; 9: 1215, 2018.
Article de Anglais | MEDLINE | ID: mdl-29951044

RÉSUMÉ

Adult T cell Leukemia/Lymphoma (ATLL) is a mature T cell malignancy associated with Human T cell Leukemia Virus type 1 (HTLV-1) infection. Among its four main clinical subtypes, the prognosis of acute and lymphoma variants remains poor. The long latency (3-6 decades) and low incidence (3-5%) of ATLL imply the involvement of viral and host factors in full-blown malignancy. Despite multiple preclinical and clinical studies, the contribution of the stromal microenvironment in ATLL development is not yet completely unraveled. The aims of this study were to investigate the role of the host microenvironment, and specifically fibroblasts, in ATLL pathogenesis and to propose a murine model for the lymphoma subtype. Here we present evidence that the oncogenic capacity of HTLV-1-immortalized C91/PL cells is enhanced when they are xenotransplanted together with human foreskin fibroblasts (HFF) in immunocompromised BALB/c Rag2-/-γc-/- mice. Moreover, cell lines derived from a developed lymphoma and their subsequent in vivo passages acquired the stable property to induce aggressive T cell lymphomas. In particular, one of these cell lines, C91/III cells, consistently induced aggressive lymphomas also in NOD/SCID/IL2Rγc KO (NSG) mice. To dissect the mechanisms linked to this enhanced tumorigenic ability, we quantified 45 soluble factors released by these cell lines and found that 21 of them, mainly pro-inflammatory cytokines and chemokines, were significantly increased in C91/III cells compared to the parental C91/PL cells. Moreover, many of the increased factors were also released by human fibroblasts and belonged to the known secretory pattern of ATLL cells. C91/PL cells co-cultured with HFF showed features reminiscent of those observed in C91/III cells, including a similar secretory pattern and a more aggressive behavior in vivo. On the whole, our data provide evidence that fibroblasts, one of the major stromal components, might enhance tumorigenesis of HTLV-1-infected and immortalized T cells, thus throwing light on the role of microenvironment contribution in ATLL pathogenesis. We also propose that the lymphoma induced in NSG mice by injection with C91/III cells represents a new murine preclinical ATLL model that could be adopted to test novel therapeutic interventions for the aggressive lymphoma subtype.

3.
Antiviral Res ; 137: 41-48, 2017 01.
Article de Anglais | MEDLINE | ID: mdl-27840202

RÉSUMÉ

Even though an estimated 10-20 million people worldwide are infected with the oncogenic retrovirus, human T-lymphotropic virus type 1 (HTLV-1), its epidemiology is poorly understood, and little effort has been made to reduce its prevalence. In response to this situation, the Global Virus Network launched a taskforce in 2014 to develop new methods of prevention and treatment of HTLV-1 infection and promote basic research. HTLV-1 is the etiological agent of two life-threatening diseases, adult T-cell leukemia and HTLV-associated myelopathy/tropical spastic paraparesis, for which no effective therapy is currently available. Although the modes of transmission of HTLV-1 resemble those of the more familiar HIV-1, routine diagnostic methods are generally unavailable to support the prevention of new infections. In the present article, the Taskforce proposes a series of actions to expand epidemiological studies; increase research on mechanisms of HTLV-1 persistence, replication and pathogenesis; discover effective treatments; and develop prophylactic and therapeutic vaccines.


Sujet(s)
Recherche biomédicale , Santé mondiale , Infections à HTLV-I , Comités consultatifs , Coûts indirects de la maladie , Infections à VIH/épidémiologie , Infections à VIH/prévention et contrôle , Infections à VIH/virologie , Infections à HTLV-I/traitement médicamenteux , Infections à HTLV-I/épidémiologie , Infections à HTLV-I/prévention et contrôle , Infections à HTLV-I/transmission , Virus T-lymphotrope humain de type 1/isolement et purification , Humains , Leucémie-lymphome à cellules T de l'adulte/virologie , Paraparésie spastique tropicale/traitement médicamenteux , Paraparésie spastique tropicale/prévention et contrôle , Paraparésie spastique tropicale/virologie , Maladies de la moelle épinière/traitement médicamenteux , Maladies de la moelle épinière/prévention et contrôle , Maladies de la moelle épinière/virologie
5.
Cancer Med ; 3(1): 1-13, 2014 Feb.
Article de Anglais | MEDLINE | ID: mdl-24402744

RÉSUMÉ

The peculiar localization of body cavity lymphomas implies a specific contribution of the intracavitary microenvironment to the pathogenesis of these tumors. In this study, primary effusion lymphoma (PEL) was used as a model of body cavity lymphoma to investigate the role of mesothelial cells, which line the serous cavities, in lymphoma progression. The crosstalk between mesothelial and lymphomatous cells was studied in cocultures of primary human mesothelial cells (HMC) with PEL cells and a xenograft mouse model of peritoneal PEL. PEL cells were found to induce type 2 epithelial-mesenchymal transition (EMT) in HMC, which converted into a myofibroblastic phenotype characterized by loss of epithelial markers (pan cytokeratin and E-cadherin), expression of EMT-associated transcriptional repressors (Snail1, Slug, Zeb1, Sip1), and acquisition of α-smooth muscle actin (α-SMA), a mesenchymal protein. A progressive thickening of serosal membranes was observed in vivo, accompanied by loss of cytokeratin staining and appearance of α-SMA-expressing cells, confirming that fibrosis occurred during intracavitary PEL development. On the other hand, HMC were found to modulate PEL cell turnover in vitro, increasing their resistance to apoptosis and proliferation. This supportive activity on PEL cells was retained after transdifferentiation, and was impaired by interferon-α2 b treatment. On the whole, our results indicate that PEL cells induce type 2 EMT in HMC, which support PEL cell growth and survival, providing a milieu favorable to lymphoma progression. Our findings provide new clues into the mechanisms involved in lymphoma progression and may indicate new targets for effective treatment of malignant effusions growing in body cavities.


Sujet(s)
Transition épithélio-mésenchymateuse/génétique , Épithélium/métabolisme , Lymphome primitif des séreuses/génétique , Protéines tumorales/biosynthèse , Animaux , Techniques de coculture , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Épithélium/anatomopathologie , Régulation de l'expression des gènes tumoraux , Humains , Lymphome primitif des séreuses/anatomopathologie , Souris , Culture de cellules primaires , Microenvironnement tumoral/génétique
6.
Blood ; 116(1): 54-62, 2010 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-20395415

RÉSUMÉ

The present study investigated the function of p13, a mitochondrial protein of human T-cell leukemia virus type 1 (HTLV-1). Although necessary for viral propagation in vivo, the mechanism of function of p13 is incompletely understood. Drawing from studies in isolated mitochondria, we analyzed the effects of p13 on mitochondrial reactive oxygen species (ROS) in transformed and primary T cells. In transformed cells (Jurkat, HeLa), p13 did not affect ROS unless the cells were subjected to glucose deprivation, which led to a p13-dependent increase in ROS and cell death. Using RNA interference we confirmed that expression of p13 also influences glucose starvation-induced cell death in the context of HTLV-1-infected cells. ROS measurements showed an increasing gradient from resting to mitogen-activated primary T cells to transformed T cells (Jurkat). Expression of p13 in primary T cells resulted in their activation, an effect that was abrogated by ROS scavengers. These findings suggest that p13 may have a distinct impact on cell turnover depending on the inherent ROS levels; in the context of the HTLV-1 propagation strategy, p13 could increase the pool of "normal" infected cells while culling cells acquiring a transformed phenotype, thus favoring lifelong persistence of the virus in the host.


Sujet(s)
Virus T-lymphotrope humain de type 1/métabolisme , Espèces réactives de l'oxygène/métabolisme , Protéines des retroviridae/métabolisme , Lymphocytes T/métabolisme , Lignée cellulaire , Cellules cultivées , Régulation de l'expression des gènes viraux , Vecteurs génétiques/génétique , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/métabolisme , Cellules HeLa , Interactions hôte-pathogène , Virus T-lymphotrope humain de type 1/physiologie , Humains , Cellules Jurkat , Lentivirus/génétique , Microscopie confocale , Mitochondries/métabolisme , Oxydoréduction , Interférence par ARN , Protéines des retroviridae/génétique , RT-PCR , Lymphocytes T/cytologie , Lymphocytes T/virologie , Transduction génétique
7.
Biochim Biophys Acta ; 1787(7): 947-54, 2009 Jul.
Article de Anglais | MEDLINE | ID: mdl-19366603

RÉSUMÉ

Human T-cell leukemia virus type-1 (HTLV-1) expresses an 87-amino acid protein named p13 that is targeted to the inner mitochondrial membrane. Previous studies showed that a synthetic peptide spanning an alpha helical domain of p13 alters mitochondrial membrane permeability to cations, resulting in swelling. The present study examined the effects of full-length p13 on isolated, energized mitochondria. Results demonstrated that p13 triggers an inward K(+) current that leads to mitochondrial swelling and confers a crescent-like morphology distinct from that caused by opening of the permeability transition pore. p13 also induces depolarization, with a matching increase in respiratory chain activity, and augments production of reactive oxygen species (ROS). These effects require an intact alpha helical domain and strictly depend on the presence of K(+) in the assay medium. The effects of p13 on ROS are mimicked by the K(+) ionophore valinomycin, while the protonophore FCCP decreases ROS, indicating that depolarization induced by K(+) vs. H(+) currents has different effects on mitochondrial ROS production, possibly because of their opposite effects on matrix pH (alkalinization and acidification, respectively). The downstream consequences of p13-induced mitochondrial K(+) permeability are likely to have an important influence on the redox state and turnover of HTLV-1-infected cells.


Sujet(s)
Virus T-lymphotrope humain de type 1 , Mitochondries/métabolisme , Protéines de transport de la membrane mitochondriale/métabolisme , Potassium/métabolisme , Espèces réactives de l'oxygène/métabolisme , Calcium/pharmacologie , ([4-(Trifluorométhoxy)phényl]hydrazono)malononitrile/pharmacologie , Virus T-lymphotrope humain de type 1/génétique , Virus T-lymphotrope humain de type 1/métabolisme , Humains , Ionophores/pharmacologie , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/ultrastructure , Pore de transition de perméabilité mitochondriale , Gonflement mitochondrial/effets des médicaments et des substances chimiques , Modèles biologiques , Perméabilité , Canaux potassiques/métabolisme , Valinomycine/pharmacologie
8.
Blood ; 113(19): 4525-33, 2009 May 07.
Article de Anglais | MEDLINE | ID: mdl-19196659

RÉSUMÉ

The peculiar site of development of primary effusion lymphoma (PEL) highlights a specific role of body cavities in the pathogenesis of this neoplasia. We used a xenograft murine model of PEL to characterize the contribution of the host microenvironment to PEL growth. The activity of a murine (ie, host-specific) interferon-alpha(1) (IFN-alpha(1))-expressing lentiviral vector (mIFN-alpha(1)-LV) was compared with that of a human (h) IFN-alpha(2)b-LV. LVs efficiently delivered the transgene to PEL cells and conferred long-term transgene expression in vitro and in vivo. Treatment of PEL-injected severe combined immunodeficiency mice with hIFN-alpha(2)b-LV significantly prolonged mice survival and reduced ascites development. Interestingly, mIFN-alpha(1)-LV showed an antineoplastic activity comparable with that observed with hIFN-alpha(2)b-LV. As mIFN-alpha(1) retained species-restricted activity in vitro, it probably acted in vivo on the intracavitary murine milieu. mIFN-alpha(1)-treated murine mesothelial cells were found to express tumor necrosis factor-related apoptosis-inducing ligand and to significantly trigger apoptosis of cocultured PEL cells in a tumor necrosis factor-related apoptosis-inducing ligand-dependent manner. These data suggest that the interaction between lymphomatous and mesothelial cells lining the body cavities may play a key role in PEL growth control and also indicate that the specific targeting of microenvironment may impair PEL development.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Vecteurs génétiques , Interféron alpha/usage thérapeutique , Lentivirus/génétique , Lymphome primitif des séreuses/traitement médicamenteux , Animaux , Cellules cultivées , Cytokines/métabolisme , Évaluation préclinique de médicament , Femelle , Humains , Interféron alpha-2 , Rein/cytologie , Rein/effets des médicaments et des substances chimiques , Rein/métabolisme , Lymphome primitif des séreuses/génétique , Lymphome primitif des séreuses/anatomopathologie , Mésoderme/cytologie , Mésoderme/effets des médicaments et des substances chimiques , Mésoderme/métabolisme , Souris , Souris SCID , Péritoine/cytologie , Péritoine/effets des médicaments et des substances chimiques , Péritoine/métabolisme , Protéines recombinantes
9.
Science ; 323(5911): 206-7, 2009 Jan 09.
Article de Anglais | MEDLINE | ID: mdl-19131607
10.
Genes Chromosomes Cancer ; 48(5): 383-96, 2009 May.
Article de Anglais | MEDLINE | ID: mdl-19170121

RÉSUMÉ

Loss of menin, a tumor suppressor coded by the MEN1 gene, is a key factor in the pathogenesis of multiple endocrine neoplasia type I and in a percentage of sporadic endocrine tumors of the pancreas and parathyroid glands. This study investigated expression of the menin protein in the normal exocrine pancreas and in pancreatic ductal adenocarcinoma (PDAC), the most common pancreatic tumor. Immunofluorescence (IF) analyses showed that menin is expressed at high levels in normal acinar and duct cells. Examination of 24 clinical samples of PDAC revealed a pronounced decrease in menin expression in all tumors examined. To identify alterations underlying this defect, we searched for disruption and epigenetic silencing of the MEN1 gene. Analysis of nine laser-microdissected tumors revealed loss of heterozygosity of intragenic (one tumor) or adjacent (three tumors) MEN1 microsatellite markers. Methylation of CpG sites in the MEN1 promoter was documented in five of 24 tumors. IF analyses also revealed low to undetectable menin expression in the PDAC cell lines MiaPaCa-2 and Panc-1. Ectopic expression of menin in these cells resulted in a marked alteration of the cell cycle, with an increase in the G1/S+G2 ratio. These findings represent the first evidence that the MEN1 gene is a target of mutation and methylation in PDAC and that menin influences the cell cycle profile of duct cells.


Sujet(s)
Carcinome du canal pancréatique/génétique , Méthylation de l'ADN , Régulation de l'expression des gènes tumoraux , Tumeurs du pancréas/génétique , Régions promotrices (génétique) , Protéines proto-oncogènes/génétique , Sujet âgé , Séquence nucléotidique , Carcinome du canal pancréatique/métabolisme , Cycle cellulaire , Lignée cellulaire tumorale , Épigenèse génétique , Femelle , Technique d'immunofluorescence , Humains , Perte d'hétérozygotie , Mâle , Adulte d'âge moyen , Données de séquences moléculaires , Analyse multifactorielle , Pancréas exocrine/cytologie , Pancréas exocrine/métabolisme , Conduits pancréatiques/cytologie , Conduits pancréatiques/métabolisme , Tumeurs du pancréas/métabolisme , Réaction de polymérisation en chaîne , Modèles des risques proportionnels , Protéines proto-oncogènes/métabolisme
11.
PLoS One ; 3(12): e4073, 2008.
Article de Anglais | MEDLINE | ID: mdl-19115001

RÉSUMÉ

Herpesvirus infection of placenta may be harmful in pregnancy leading to disorders in fetal growth, premature delivery, miscarriage, or major congenital abnormalities. Although a correlation between human herpesvirus 8 (HHV-8) infection and abortion or low birth weight in children has been suggested, and rare cases of in utero or perinatal HHV-8 transmission have been documented, no direct evidence of HHV-8 infection of placenta has yet been reported. The aim of this study was to evaluate the in vitro and in vivo susceptibility of placental cells to HHV-8 infection. Short-term infection assays were performed on placental chorionic villi isolated from term placentae. Qualitative and quantitative HHV-8 detection were performed by PCR and real-time PCR, and HHV-8 proteins were analyzed by immunohistochemistry. Term placenta samples from HHV-8-seropositive women were analyzed for the presence of HHV-8 DNA and antigens. In vitro infected histocultures showed increasing amounts of HHV-8 DNA in tissues and supernatants; cyto- and syncitiotrophoblasts, as well as endothelial cells, expressed latent and lytic viral antigens. Increased apoptotic phenomena were visualized by the terminal deoxynucleotidyl transferase-mediated deoxyuridine nick end-labeling method in infected histocultures. Ex vivo, HHV-8 DNA and a latent viral antigen were detected in placenta samples from HHV-8-seropositive women. These findings demonstrate that HHV-8, like other human herpesviruses, may infect placental cells in vitro and in vivo, thus providing evidence that this phenomenon might influence vertical transmission and pregnancy outcome in HHV-8-infected women.


Sujet(s)
Infections à Herpesviridae/virologie , Herpèsvirus humain de type 8/isolement et purification , Maladies du placenta/virologie , Placenta/virologie , Apoptose , ADN viral/métabolisme , Cellules endothéliales/métabolisme , Femelle , Infections à Herpesviridae/immunologie , Herpèsvirus humain de type 8/génétique , Herpèsvirus humain de type 8/immunologie , Humains , Immunohistochimie , Placenta/immunologie , Maladies du placenta/immunologie , Grossesse , Trophoblastes/métabolisme
12.
Cancer Res ; 67(18): 8605-14, 2007 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-17875700

RÉSUMÉ

The chemokine receptor CXCR4 plays a central role in organ-specific homing and tumor spreading and is induced by hypoxia. B lymphocytes are exposed to low oxygen tensions during their development, but the influence of hypoxia on their physiology is poorly understood. Here, we show that hypoxia is associated with up-regulation of CXCR4 expression in human normal and malignant B cells, through both transcriptional and posttranslational mechanisms. However, a dichotomic functional response to CXCR4 triggering was observed: both peripheral B cells and lymphomas arising from mature B cells displayed increased responses to CXCR4 triggering under hypoxia, whereas germinal center (GC) B cells as well as GC-derived lymphomas showed CXCR4 receptor desensitization. This phenomenon was associated with differential modulation of key signal-transducing molecules, including mitogen-activated protein kinase phosphatase-1 and regulator of G protein signaling molecule-1. The unresponsiveness of GC-derived lymphomatous B cells to CXCR4 triggering under hypoxia may have implications for the development and pathogenesis of GC-derived lymphoid tumors.


Sujet(s)
Lymphocytes B/métabolisme , Lymphocytes B/physiologie , Lymphome B/métabolisme , Lymphome B/anatomopathologie , Récepteurs CXCR4/biosynthèse , Animaux , Lymphocytes B/anatomopathologie , Lymphome de Burkitt/génétique , Lymphome de Burkitt/métabolisme , Lymphome de Burkitt/anatomopathologie , Hypoxie cellulaire/physiologie , Lignée cellulaire tumorale , Dual Specificity Phosphatase 1/biosynthèse , Dual Specificity Phosphatase 1/génétique , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Lymphome B/génétique , Souris , Souris SCID , Protéines RGS/biosynthèse , Protéines RGS/génétique , Petit ARN interférent/génétique , Récepteurs CXCR4/génétique , Transcription génétique , Régulation positive
13.
J Immunol ; 178(2): 1122-35, 2007 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-17202376

RÉSUMÉ

IFNs are highly pleiotropic cytokines also endowed with marked antiangiogenic activity. In this study, the mRNA expression profiles of endothelial cells (EC) exposed in vitro to IFN-alpha, IFN-beta, or IFN-gamma were determined. We found that in HUVEC as well as in other EC types 175 genes were up-regulated (>2-fold increase) by IFNs, including genes involved in the host response to RNA viruses, inflammation, and apoptosis. Interestingly, 41 genes showed a >5-fold higher induction by IFN-alpha in EC compared with human fibroblasts; among them, the gene encoding the angiostatic chemokine CXCL11 was selectively induced by IFN-alpha in EC along with other genes associated with angiogenesis regulation, including CXCL10, TRAIL, and guanylate-binding protein 1. These transcriptional changes were confirmed and extended by quantitative PCR analysis and ELISA; whereas IFN-alpha and IFN-beta exerted virtually identical effects on transcriptome modulation, a differential gene regulation by type I and type II IFN emerged, especially as far as quantitative aspects were concerned. In vivo, IFN-alpha-producing tumors overexpressed murine CXCL10 and CXCL11, guanylate-binding protein 1, and TRAIL, with evidence of CXCL11 production by tumor-associated EC. Overall, these findings improve our understanding of the antiangiogenic effects of IFNs by showing that these cytokines trigger an antiangiogenic transcriptional program in EC. Moreover, we suggest that quantitative differences in the magnitude of the transcriptional activation of IFN-responsive genes could form the basis for cell-specific transcriptional signatures.


Sujet(s)
Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/génétique , Interférons/pharmacologie , Animaux , Cellules cultivées , Chimiokine CXCL10 , Chimiokine CXCL11 , Chimiokines CXC/métabolisme , Expression des gènes , Analyse de profil d'expression de gènes , Humains , Cinétique , Mâle , Souris , Séquençage par oligonucléotides en batterie , ARN messager/génétique , Récepteurs CXCR3 , Récepteurs aux chimiokines/métabolisme , Peau/vascularisation , Peau/effets des médicaments et des substances chimiques , Peau/métabolisme , Ligand TRAIL/métabolisme , Cordon ombilical/métabolisme
14.
J Clin Microbiol ; 44(11): 3863-71, 2006 Nov.
Article de Anglais | MEDLINE | ID: mdl-16943357

RÉSUMÉ

To investigate the impact of pregnancy on human herpesvirus 8 (HHV-8) reactivation in human immunodeficiency virus type 1 (HIV-1)-infected women, the HHV-8 DNA presence and load were analyzed in peripheral blood mononuclear cells (PBMCs) and cervicovaginal secretions (CVSs) from 15 pregnant women coinfected with HIV-1 and HHV-8. HHV-8 detection was analyzed in relation to anti-HHV-8 antibodies and HIV-1-related parameters. Nucleotide sequence analysis of an ORFK1 hypervariable region of the HHV-8 strains was performed. HHV-8 was detected in maternal PBMCs (5/15 women) from the second trimester and in CVSs (5/15 women) mainly from the third trimester. The HHV-8 load significantly increased late in pregnancy in both maternal compartments and was associated with a significant increase in HIV-1 shedding in the genital tract. Antilytic antibodies were significantly more common in HHV-8 DNA-positive women. An elevated HHV-8 load was found in the PBMCs of an infant born to a mother with large amounts of HHV-8 in both compartments at delivery. Different ORFK1 subtypes were found in maternal samples, whereas the same subtype was identified in the mother-child pair. These data suggest that pregnancy may induce HHV-8 replication in HIV-1-infected women. An augmented HHV-8 load may, in turn, influence mother-to-child transmission, since one of the HIV-1-infected mothers with HHV-8 reactivation transmitted her ORFK1 subtype to the infant, who showed a high level of HHV-8 viremia indicative of a primary infection. This finding documents for the first time the perinatal transmission of a specific HHV-8 subtype. Vertical transmission may thus play a role in HHV-8 spread also in areas of subendemicity among HIV-1-infected women.


Sujet(s)
Syndrome d'immunodéficience acquise/virologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Herpèsvirus humain de type 8/isolement et purification , Complications infectieuses de la grossesse/virologie , Activation virale , Adulte , Séquence d'acides aminés , Séquence nucléotidique , ADN viral/sang , Femelle , Herpèsvirus humain de type 8/physiologie , Humains , Nouveau-né , Agranulocytes/virologie , Données de séquences moléculaires , Réaction de polymérisation en chaîne , Grossesse , Excrétion virale
15.
Eur J Cancer ; 42(10): 1475-83, 2006 Jul.
Article de Anglais | MEDLINE | ID: mdl-16759848

RÉSUMÉ

The BRCA1 gene is responsible for a high number of hereditary breast and ovarian cancers that cluster in families with a strong genetic predisposition. Despite intense investigation, the accumulating findings on BRCA1 biological functions have not yet been translated into specific therapeutic approaches, also due to the lack of suitable experimental models. The purpose of this study was to establish and characterize cell cultures and xenografts from patients with BRCA1 -/- ovarian cancers. We derived two ovarian cancer cell lines, termed PD-OVCA1 and PD-OVCA2, both from patients previously treated with chemotherapy, that propagate in SCID mice as well as in vitro for a limited number of passages. Both cell lines expressed cytokeratins and the CA125 tumour marker. A detailed molecular characterization highlighted both constitutive and somatic genetic events that abrogate BRCA1 gene function. Both cell lines were shown to lose the wild type BRCA1 allele; intriguingly, these deletions were apparently accompanied by gain of one or more copies of the mutant alleles. Finally, a genomic profile of major chromosomal aberrations was obtained by the Multiplex Ligation-dependent Probe Amplification (MLPA) technique, which disclosed chromosomal imbalances targeting specific genes in each cell line. The PD-OVCA1 and PD-OVCA2 ovarian cancer cell lines will provide a valuable tool for new experimental models for the study of BRCA1-associated tumour biology.


Sujet(s)
Gène BRCA1 , Tumeurs de l'ovaire/anatomopathologie , Cellules cancéreuses en culture/anatomopathologie , Sujet âgé , Animaux , Antigènes CA-125/métabolisme , Techniques de culture cellulaire/méthodes , Femelle , Génome , Génotype , Humains , Souris , Tumeurs de l'ovaire/génétique , RT-PCR , Transplantation hétérologue/méthodes , Cellules cancéreuses en culture/métabolisme
17.
J Med Virol ; 78(7): 933-7, 2006 Jul.
Article de Anglais | MEDLINE | ID: mdl-16721860

RÉSUMÉ

Patterns of endemicity of human herpesvirus 8 (HHV8) are still undefined in some European populations, such as those from Western Balkan countries. Serum samples from 605 human immunodeficiency virus-seronegative subjects (299 Albanians and 306 Kosovars) were tested for the presence of HHV8 antibodies to a capsid-related open reading frame (ORF65)-encoded protein and a latency-associated nuclear antigen (LANA) to determine HHV8 seroprevalence in populations from Albania and from the Kosovo region of former Yugoslavia. Levels of co- circulation with hepatitis A (HAV) and hepatitis B (HBV) viruses were also determined. HHV8 antibodies to at least one of the two antigens were detected in 28.8% of Albanians and 18% of Kosovars. The seroprevalence of HHV8 was found to be 25.0 and 16.8% in Albanian and Kosovar children (

Sujet(s)
Infections à Herpesviridae/épidémiologie , Herpèsvirus humain de type 8 , Adolescent , Adulte , Albanie/épidémiologie , Anticorps antiviraux/sang , Enfant , Enfant d'âge préscolaire , Femelle , Hépatite A/complications , Hépatite B/complications , Infections à Herpesviridae/complications , Infections à Herpesviridae/immunologie , Herpèsvirus humain de type 8/immunologie , Humains , Nourrisson , Mâle , Adulte d'âge moyen , Études séroépidémiologiques , Protéines virales/immunologie , Yougoslavie/épidémiologie
18.
Proc Natl Acad Sci U S A ; 103(11): 4216-21, 2006 Mar 14.
Article de Anglais | MEDLINE | ID: mdl-16537511

RÉSUMÉ

Tumor growth is currently viewed as a phenomenon associated with neovascularization and sustained production of angiogenic factors, but whether a transient angiogenic switch may trigger tumor growth remains unclear. Here, we report that leukemia cells (MOLT-3) were poorly angiogenic and remained dormant when injected s.c. into immunodeficient mice. However, progressive growth of lymphoid tumors was invariably recorded when irradiated angiogenic cells from Kaposi's sarcoma (KS-IMM) were locally coinjected with MOLT-3 cells or administered later. The persistence of KS-IMM cells in vivo was tracked by flow cytometry and real-time PCR analysis, and it was limited to a few days, during which angiogenesis was induced and preceded tumor growth. The engraftment of other types of poorly tumorigenic cancer cells was also greatly improved by irradiated KS-IMM cells. Moreover, short-term treatment with angiogenic factors, including basic FGF or VEGF, either given as recombinant factors or delivered by retroviral vectors, also accelerated tumor growth. These findings may emphasize that tumor angiogenesis is a process requiring a higher amount of angiogenic factors for its induction than maintenance.


Sujet(s)
Tumeurs expérimentales/vascularisation , Néovascularisation pathologique/étiologie , Sarcome de Kaposi/vascularisation , Animaux , Lignée cellulaire tumorale , Facteur de croissance fibroblastique de type 2/pharmacologie , Humains , Souris , Souris de lignée NOD , Souris SCID , Transplantation tumorale , Protéines recombinantes/pharmacologie , Facteurs temps , Transplantation hétérologue , Facteur de croissance endothéliale vasculaire de type A/pharmacologie
19.
Exp Cell Res ; 312(8): 1390-400, 2006 May 01.
Article de Anglais | MEDLINE | ID: mdl-16483570

RÉSUMÉ

Defects in the regulation of programmed cell death play a fundamental role in the development of neoplasia and neurological disorders, both of which are linked to the human T-cell leukemia/lymphoma virus type 1 (HTLV-1) infection. We previously showed that the HTLV-1 Tax protein protects from apoptosis induced by serum starvation by preventing cytochrome c release and Bax relocation to mitochondria, two early events in the mitochondrial apoptotic pathway. As a natural extension of these findings, and to better define the action of Tax, in the present study, we investigated the outcome of Tax and two mutants which are inactive in CREB/ATF (M47) or NF-kappaB (M22) pathways, in the control of apoptosis induced by the proapoptotic Bax protein. We found that activation of CREB, rather than NF-kappaB, is a key phenomenon in preventing apoptosis. Furthermore, the importance of CREB activation is strengthened by experiments with CREB mutants, treatment with forskolin, and in situ analysis of P-CREB status in cells transfected with Tax or its nonprotecting M47 mutant. Considered together, these results underscore a primary role of CREB in preventing apoptosis triggered by Bax, and suggest that Tax might act by affecting the phosphorylation state of CREB.


Sujet(s)
Apoptose/physiologie , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Produits du gène tax/métabolisme , Infections à HTLV-I/métabolisme , Virus T-lymphotrope humain de type 1/métabolisme , Facteurs de transcription/physiologie , Apoptose/génétique , Colforsine/pharmacologie , Protéine de liaison à l'élément de réponse à l'AMP cyclique/génétique , Régulation de l'expression des gènes viraux/physiologie , Produits du gène tax/génétique , Infections à HTLV-I/génétique , Infections à HTLV-I/physiopathologie , Cellules HeLa , Virus T-lymphotrope humain de type 1/génétique , Humains , Mutation/physiologie , Facteur de transcription NF-kappa B/génétique , Facteur de transcription NF-kappa B/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Facteurs de transcription/effets des médicaments et des substances chimiques , Activation de la transcription/effets des médicaments et des substances chimiques , Activation de la transcription/physiologie , Protéine Bax/génétique , Protéine Bax/métabolisme
20.
Hum Gene Ther ; 16(8): 957-70, 2005 Aug.
Article de Anglais | MEDLINE | ID: mdl-16076254

RÉSUMÉ

Ovarian cancer represents a suitable disease for gene therapy because of the containment of neoplastic cells in the peritoneal cavity even at advanced tumor stages. The aim of this study was to investigate whether intraperitoneal administration of a lentiviral vector encoding murine interferon-alpha (LV-IFN) could have therapeutic activity in a transplantable ovarian cancer model. Multiple injections of low amounts of LV-IFN into severe combined immunodeficiency (SCID) mice bearing IGROV-1 or OC316 ovarian cancer cells elicited remarkable antitumor activity, leading to prolongation of survival in the majority of animals. A definitive cure was obtained in animals bearing PD-OVA#1 tumors, generated by injecting tumor cells isolated from the ascitic fluid of a patient into SCID mice. Interferon-alpha levels were detected in the peritoneal fluids but not in the serum of treated mice, indicating that production of the cytokine is mainly local, by both tumor and normal cells of the host. Antitumor effects were associated with a remarkable decrease in the formation of hemorrhagic ascites, an increase in ischemic tumor necrosis, and a reduction in microvessel density. In conclusion, our findings show that intracavitary IFN-alpha gene therapy, using a lentiviral vector, provides strong antitumor effects in murine models of ovarian cancer and reinforces the evidence that angiogenesis inhibition is a promising strategy for the treatment of localized tumors.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Techniques de transfert de gènes , Thérapie génétique/méthodes , Interféron alpha/génétique , Interféron alpha/usage thérapeutique , Néovascularisation pathologique , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Animaux , Antinéoplasiques/administration et posologie , Antinéoplasiques/pharmacocinétique , Modèles animaux de maladie humaine , Femelle , Vecteurs génétiques , Humains , Perfusions parentérales , Interféron alpha/administration et posologie , Interféron alpha/pharmacocinétique , Lentivirus/génétique , Souris , Souris SCID , Tumeurs de l'ovaire/médecine vétérinaire , Survie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...