938 resultados para Transfection
Resumo:
Islet-brain-1 (IB1)/c-Jun N-terminal kinase interacting protein 1 (JIP-1) is a scaffold protein that is expressed at high levels in neurons and the endocrine pancreas. IB1/JIP-1 interacts with the c-Jun N-terminal kinase and mediates the specific physiological stimuli (such as cytokines). However, the potential role of the protein in the pituitary has not been evaluated. Herein, we examined expression of the gene encoding IB1/JIP-1 and its translated product in the anterior pituitary gland and a pituitary cell line, GH3. We then examined the potential role of IB1/JIP-1 in controlling TSH-beta gene expression. Exposure of GH3 cells to TRH stimulated the expression of IB1/JIP-1 protein levels, mRNA, and transcription of the promoter. The increase of IB1/JIP-1 content by transient transfection study of a vector encoding IB1/JIP-1 or by the stimulation of TRH stimulates TSH-beta promoter activity. This effect is not found in the presence of a mutated nonfunctional (IB1S59N) IB1/JIP-1 protein. Together, these facts point to a central role of the IB1/JIP-1 protein in the control of TRH-mediated TSH-beta stimulation.
Resumo:
PURPOSE: We have investigated the expression and regulation of 15-hydroxyprostaglandin dehydrogenase (15-PGDH) in gastric cancer. EXPERIMENTAL DESIGN: Clinical gastric adenocarcinoma samples were analyzed by immunohistochemistry and quantitative real-time PCR for protein and mRNA expression of 15-PGDH and for methylation status of 15-PGDH promoter. The effects of interleukin-1beta (IL-1beta) and epigenetic mechanisms on 15-PGDH regulation were assessed in gastric cancer cell lines. RESULTS: In a gastric cancer cell line with a very low 15-PGDH expression (TMK-1), the 15-PGDH promoter was methylated and treatment with a demethylating agent 5-aza-2'-deoxycytidine restored 15-PGDH expression. In a cell line with a relatively high basal level of 15-PGDH (MKN-28), IL-1beta repressed expression of 15-PGDH mRNA and protein. This effect of IL-1beta was at least in part attributed to inhibition of 15-PGDH promoter activity. SiRNA-mediated knockdown of 15-PGDH resulted in strong increase of prostaglandin E(2) production in MKN-28 cells and increased cell growth of these cells by 31% in anchorage-independent conditions. In clinical gastric adenocarcinoma specimens, 15-PGDH mRNA levels were 5-fold lower in gastric cancer samples when compared with paired nonneoplastic tissues (n = 26) and 15-PGDH protein was lost in 65% of gastric adenocarcinomas (n = 210). CONCLUSIONS: 15-PGDH is down-regulated in gastric cancer, which could potentially lead to accelerated tumor progression. Importantly, our data indicate that a proinflammatory cytokine linked to gastric carcinogenesis, IL-1beta, suppresses 15-PGDH expression at least partially by inhibiting promoter activity of the 15-PGDH gene.
Resumo:
The orphan receptor CRF2-4 is a member of the class II cytokine receptor family (CRF2), which includes the interferon receptors, the interleukin (IL) 10 receptor, and tissue factor. CRFB4, the gene encoding CRF2-4, is located within a gene cluster on human chromosome 21 that comprises three interferon receptor subunits. To elucidate the role of CRF2-4, we disrupted the CRFB4 gene in mice by means of homologous recombination. Mice lacking CRF2-4 show no overt abnormalities, grow normally, and are fertile. CRF2-4 deficient cells are normally responsive to type I and type II interferons, but lack responsiveness to IL-10. By approximately 12 wk of age, the majority of mutant mice raised in a conventional facility developed a chronic colitis and splenomegaly. Thus, CRFB4 mutant mice recapitulate the phenotype of IL-10-deficient mice. These findings suggest that CRF2-4 is essential for IL-10-mediated effects and is a subunit of the IL-10 receptor.
Resumo:
Purpose:We analyzed the transcriptional activity of disease-causing NR2E3 mutant proteins in a heterologous system. NR2E3 belongs to the nuclear receptor superfamily of transcription factors, characterized by evolutionary-conserved DNA-binding (DBD) and ligand-binding (LBD) domains. NR2E3 acts in concert with the transcription factors CRX and NRL to repress cone-specific genes and activate rod-specific genes in rod photoreceptors. During development, NR2E3 is also required to suppress cone cell generation from retinal progenitor cells. In humans, mutations in NR2E3 have been associated with the recessively inherited enhanced short wavelength sensitive (S-) cone syndrome (ESCS), the Goldman-Favre syndrome, and, more recently, with autosomal dominant retinitis pigmentosa (adRP). Methods:The different NR2E3 mutants were generated by QuickChangeR mutagenesis and analyzed by transfection in heterologous HEK293T cells. Results:In transactivation assays in HEK293T cells, the adRP-linked p.G56R mutant protein exhibited a more severe effect both in activation of a rhodopsin promoter reporter construct and in repression of M-opsin promoter reporter construct, than the ESCS-linked R76Q, R76W, G88V, R97H, R104Q, R104W mutants of the DBD. In contrast, the ESCS-linked p.R311Q mutant of the LBD behaved like the NR2E3 wild-type protein in these assays. By co-expressing the corepressors atrophin-1 and -2, a differential repression of the M-opsin promoter was observed in presence of the p.R311Q, p.R385P and p.M407K. Interestingly, corepressor expression also affected the activity of CRX, but not NRL, in both rhodopsin and M-opsin transactivation assays. Conclusions:Taken together, these in vitro results suggest a distinct disease mechanism for the adRP-linked mutation, but open the possibility of different mechanisms for the development of ESCS that is clinically characterized by important phenotypic variations.
Resumo:
Islet-brain 1 (IB1) is the human and rat homologue of JIP-1, a scaffold protein interacting with the c-Jun amino-terminal kinase (JNK). IB1 expression is mostly restricted to the endocrine pancreas and to the central nervous system. Herein, we explored the transcriptional mechanism responsible for this preferential islet and neuronal expression of IB1. A 731-bp fragment of the 5' regulatory region of the human MAPK8IP1 gene was isolated from a human BAC library and cloned upstream of a luciferase reporter gene. This construct drove high transcriptional activity in both insulin-secreting and neuron-like cells but not in unrelated cell lines. Sequence analysis of this promoter region revealed the presence of a neuron-restrictive silencer element (NRSE) known to bind repressor zinc finger protein REST. This factor is not expressed in insulin-secreting and neuron-like cells. By mobility shift assay, we confirmed that REST binds to the NRSE present in the IB1 promoter. Once transiently transfected in beta-cell lines, the expression vector encoding REST repressed IB1 transcriptional activity. The introduction of a mutated NRSE in the 5' regulating region of the IB1 gene abolished the repression activity driven by REST in insulin-secreting beta cells and relieved the low transcriptional activity of IB1 observed in unrelated cells. Moreover, transfection in non-beta and nonneuronal cell lines of an expression vector encoding REST lacking its transcriptional repression domain relieved IB1 promoter activity. Last, the REST-mediated repression of IB1 could be abolished by trichostatin A, indicating that deacetylase activity is required to allow REST repression. Taken together, these data establish a critical role for REST in the control of the tissue-specific expression of the human IB1 gene.
Resumo:
Introduction: Glioblastoma (WHO Grade IV glioma) is the most frequent and most¦malignant primary tumor of the brain. With a mean survival of 15 months despite¦multidisciplinary management combining surgery, chemo- and radiotherapy, the prognosis¦is poor. Different studies measured a down-regulation of Wnt Inhibitory Factor 1 (WIF1)¦expression in a majority of gliobastoma due to genetic and epigenetic regulation. Recently,¦a focus on chromosome 12 identified WIF1 as a potential tumor suppressor gene. In¦previous results, transfected glioblastoma cells with ectopic expression of WIF1 had a¦decreased growth rate and adopted a senescence-like phenotype. In this report, we first¦investigated the effect of WIF1 re-expression in glioblastoma cell lines to see if Wnt¦inhibition by WIF1 can lead to senescence. To look further, we assessed p21 and c-Myc¦expression. p21 has a key role in senescence onset and is directly inhibited by c-Myc,¦itself a target of Wnt-pathway. We thus looked if a variation of expression of these genes is¦triggered by WIF1 activity. Finally, as autophagy is thought to play a role in senescence¦onset, we analyzed the expression of different autophagy genes. We therefore looked for¦an association between autophagy activity and senescent phenotype in WIF1-¦overexpressing cell lines.¦Methods: WIF1-overexpressing clones were selected after transfection of stable¦glioblastoma cell lines. Analysis were made through quantitative Polymerase Chain¦Reaction (qPCR), Fluorescence-activated Cell Sorting (FACS) and histochemistry.¦IGFBP7 and ALDH1A3 have been selected to reflect senescence. ATG5, ATG7 and ULK3¦have been selected to reflect autophagy activity.¦Results: Using FACS analysis, we found a higher percentage of large cells with increased¦granularity amongst WIF1-overexpressing cell lines, which are characteristics of¦senescence. In addition, histochemistry showed a higher percentage of multi-nucleated,¦beta-galactosidase positive cells in the same cell lines. An increased expression of genes¦associated with senescence was found as well. All characteristics were correlated with¦levels of WIF1 expression. We did not find any association between p21 and WIF1¦expression. No correlation between WIF1 and c-Myc expression was noticed either. In one¦of the two cell lines analyzed, the expression of autophagy genes showed some¦correlation with expression of WIF1 and expression of genes associated with senescence.¦Discussion: After investigations and characterizations on multiple levels, we have¦evidence for a senescence phenotype upon WIF1-overexpressing cell lines. This gives a¦role to Wnt pathway in the tumorigenicity of glioblastoma. Further experiments are¦required to investigate how Wnt inhibition leads to senescence. The role of autophagy in¦our senescent cells is here still unclear. Some correlations can be found, letting us think¦that there is indeed some involvement of autophagy. However, it is yet to soon to explain¦this relationship. Further experiments are required again to confirm the preliminary results¦and analyze the variations of autophagy activity within time.
Resumo:
RESUME Nous avons étudié le rôle de deux molécules, le Transfon-ning Growth Factor (TGF-β) et l'oxyde nitrique (NO), dans le processus métastatique. Deux clones tumoraux ont été sélectionnés à partir d'un carcinome du côlon pour leur différence de potentiel tumorigénique dans des rats syngéniques. La croissance tumorale du clone progressif PROb a été corrélée à sa capacité à sécréter le TGF-β actif Cependant, la transfection du clone régressif REGb, sécrétant du TGF-β latent, par une vecteur codant pour le TGF-β bio-actif n'a pas permis d'induire le développement tumoral. Les deux clones tumoraux présentent des activités des protéases MMP-2, APN et DPPIV identiques et qui ne semblent pas modifiées par le TGF-β. L'interaction des cellules tumorales avec l'endothélium et l'activité de la NO synthase (iNOS) responsable de la synthèse de NO sont impliqués dans la progression de nombreux cancers. Le clone PROb, mais pas le clone REGb, inhibe l'activation de la iNOS des cellules endothéliales par sa sécrétion de TGF-β actif Les deux clones montrent cependant des propriétés d'adhésion identiques aux cellules endothéliales et sont capables d'inhiber par contact cellulaire direct l'activation de la iNOS endothéliale. Ceci suggère que ces contacts directs pourraient créer un micro-environnement favorable à la conversion du TGF-β latent en TGF-β actif ou à d'autres interactions moléculaires pouvant réguler l'activation endothéliale. Par ailleurs, les deux clones activent des macrophages du système nerveux central, organe où ils ne forment pas de métastases, mais pas les macrophages circulants, illustrant des mécanismes différentiels et spécifiques dans l'activation de différents types de cellules immunitaires. Afin de mieux comprendre le rôle du NO dans la dissémination métastatique, deux clones cellulaires différant par le taux d'activité de la iNOS ont été sélectionnés à partir de la lignée murine parentale de carcinome du sein EMT-6. Bien que le NO soit un inhibiteur potentiel de la prolifération cellulaire, les deux clones montrent des propriétés prolifératives identiques in vitro. Les cellules EMT-6H qui produisent peu de NO in vitro forment de nombreux nodules tumoraux pulmonaires in vivo corrélés à une mortalité significative des souris syngéniques injectées. Les cellules EMT-6J qui présentent une expression élevée de iNOS et de NO induisent de rares nodules tumoraux pulmonaires et peu de mortalité. Dans ce modèle, l'expression tumorale de NO semble donc défavoriser la croissance tumorale. Les deux clones cellulaires ont des propriétés identiques d'adhésion et de prolifération mesurées in vitro sur des cellules endothéliales primaires isolées de différents organes et in vivo par une colocalisation identique dans les poumons de souris syngéniques 48h après leur injection. Les cellules EMT-6H présentent une activité MMP-2 plus élevée alors que les activités des protéases APN et DPPIV sont identiques dans les deux clones cellulaires. Le TGF-β soluble ainsi que les fibroblastes primaires bloquent la prolifération des deux clones cellulaires. Cependant, l'activation préalable des fibroblastes par du TGF-β restaure partiellement la prolifération du clone EMT-6H mais pas celle du clone EMT-6J. Ces résultats montrent que le rôle de molécules telles que le TGF-β et le NO tumoral dans la progression tumorale doit être considéré dans un contexte d'interactions des cellules tumorales avec les différentes types cellulaires de l'hôte: en particulier, notre travail souligne que les macrophages et les fibroblastes sont déterminants dans la progression métastatique des carcinomes du côlon ou du sein. RESUME DESTINE A UN LARGE PUBLIC Les métastases tumorales, disséminées et intraitables par chirurgie, représentent un problème majeur dans le traitement clinique du cancer. Elles sont dues à des cellules tumorales qui ont migré de leur site tumoral primaire, circulé et survécu dans le système vasculaire de l'hôte, échappé au système immunitaire, adhéré à et survécu sur l'endothélium des vaisseaux, et envahi le tissu sous-jacent où elles ont proliféré. Cette capacité à former des métastases implique de nombreux facteurs dont certains ont été identifiés mais dont le rôle reste controversé dans les différentes études. Nous nous sommes intéressés au rôle de l'oxyde nitrique (NO) et du facteur de croissance et de transformation cellulaire TGF-β. Dans les carcinomes du sein, l'expression des enzymes responsables de la synthèse de NO a été corrélée avec l'invasion tumorale mais aussi avec un pronostic favorable selon les études. Deux clones cellulaires ont été isolés à partir de la tumeur mammaire EMT-6 chez la souris. Le clone EMT-6H sécrète peu de NO et forme de nombreuses tumeurs dans les poumons des souris *entraînant leur décès. Le clone EMT-6J sécrète beaucoup de NO et ne se développe que peu dans les poumons. Dans ce modèle expérimental, le NO semble donc défavoriser la croissance tumorale. L'analyse des interactions avec les cellules de l'hôte rencontrées lors de la formation de métastases pulmonaires a montré que les deux clones cellulaires adhérent et prolifèrent de manière similaire sur les cellules endothéliales tapissant l'intérieur des vaisseaux sanguins. L'arrêt des cellules tumorales dans les poumons ne permet donc pas d'expliquer la différence de croissance tumorale. Cependant, le clone agressif EMT-6H présente une activité élevée d'une protéase (MMP-2) qui lui permettrait par la suite d'envahir le tissu pulmonaire. Par ailleurs, l'activation des fibroblastes du tissu pulmonaire par le TGF-β, une molécule observée dans des conditions inflammatoires, permet au clone agressif EMT-6H de proliférer mais inhibe la croissance du clone EMT-6J. Dans un modèle expérimental de carcinome du côlon, le TGF-β est considéré favorable à la croissance tumorale. Isolées à partir de la même tumeur initiale, deux lignées de cellules ont des comportements opposés lorsqu'elles sont injectées sous la peau des rats. La capacité de la lignée PROb à former des tumeurs a été corrélée à la sécrétion de TGF-β actif L'introduction du gène codant pour le TGF-β actif dans la lignée REGb, qui ne sécrète pas de TGF-β actif et ne forme pas de tumeurs chez le rat, ne restaure pas leur potentiel tumorigénique. Dans ce modèle, l'expression de TGF-β actif ne semble donc pas suffisante à la croissance tumorale. Les interactions avec différents types cellulaires de l'hôte ont été étudiées. Les deux lignées tumorales adhérent de manière similaire sur les cellules endothéliales et sont capables d'inhiber leur activation, un mécanisme qui pourrait participer à la destruction. Les deux lignées activent les cellules immunitaires du système nerveux central, un organe où elles ne forment pas de métastase. Ces résultats suggèrent que la sélection des cellules métastatiques ne s'effectue pas sur l'endothélium des vaisseaux sanguins mais à des étapes ultérieures dans le micro- environnement cellulaire du nouvel organe colonisé. SUMMARY Metastasis results from the migration of tumor cells from their primary tumor, circulation through the bloodstream, attachment to the endothelium, and invasion of the surrounding tissue where they create a microenvironnement favoring their growth. This multistep process implies various cellular interactions and molecules. Among those, we were interested in the role of the Transforming Growth Factor beta (TGF-β) and the nitric oxide (NO). Two cell lines were isolated from a rat colon tumor and assessed for their metastatic potential in vivo. The PROb cell line that expresses active TGF-β formed subcutaneous tumors in rats while the REGb cell line that expresses only latent TGF-β did not. Transfection of REGb cells with a plasmid encoding for the active form of TGF-β failed to restore their metastatic ability. Thus TGF-β secretion is not sufficient to induce colon carcinoma progression. Activities of various proteases such as APN, DPPIV and MMP were similar in both cell lines and were not regulated by TGF-β. Interactions with the endothelium as well as NO synthase activity (iNOS) and local NO concentrations are believed to be crucial steps in cancer metastasis. Coculture of the two clones with endothelial cells inhibited the cytokine-triggered activation of the iNOS enzyme in primary rat endothelial cells but only PROb cells were capable of increasing the expression of IL-6, a protumoral interleukin that may participate in the impairment of the anti-tumoral immune response of the host. Both cell lines exhibited potential to activate microglial cells but not bone marrow-derived macrophages, pointing to a differential regulation of specialized immune cells. To better understand the conflicting role of NO in breast cancer progression, two cell clones were selected from the murine tumorigenic cell line EMT-6 based on their iNOS activity and NO secretion. Although NO has been shown to inhibit cell proliferation, the two cell clones exhibited similar proliferation rates in vitro. The EMT-6H cells expressed little NO and grew actively in the lungs of syngenic mice, leading to their death. Opposite results were observed with the EMT-6J cells. In these in vivo conditions, NO seems to impair tumor growth. Both clones exhibited similar in vitro adhesive properties to primary endothelial cells isolated from various mouse organs and similar localization in the lungs of mice 48 hours after injection. Sustained metalloproteinase MMP-2 activity was detected in the tumorigenic EMT-6H clone, but not in the EMT-6J cells while other proteases such as APN and DPPIV showed no difference. These results suggested that the two clones differed in invasion steps following adhesion to the endothelium and that NO did not participate in previous steps. Consistent with this, both soluble TGF-β and supernatants of cultures of mouse primary lung fibroblasts inhibited the growth of the two clones. However, previous activation of these fibroblasts with TGF-β restored the growth of the tumorigenic EMT-6H cells, but not of EMT-6J cells. Altogether, these results indicate that the role of a given molecule, such as NO or TGF-β, must be considered in a context of interaction of tumor cells with host cells. They further imply that interaction of tumor cells with specialized immune cells and with stromal cells of the colonized organ, rather than with the endothelium, are critical in regulating metastasis.
Resumo:
Fas, a death domain-containing member of the tumor necrosis factor receptor family and its ligand FasL have been predominantly studied with respect to their capability to induce cell death. However, a few studies indicate a proliferation-inducing signaling activity of these molecules too. We describe here a novel signaling pathway of FasL and the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) that triggers transcriptional activation of the proto-oncogene c-fos, a typical target gene of mitogenic pathways. FasL- and TRAIL-mediated up-regulation of c-Fos was completely dependent on the presence of Fas-associated death domain protein (FADD) and caspase-8, but caspase activity seemed to be dispensable as a pan inhibitor of caspases had no inhibitory effect. Upon overexpression of the long splice form of cellular FADD-like interleukin-1-converting enzyme (FLICE) inhibitory protein (cFLIP) in Jurkat cells, FasL- and TRAIL-induced up-regulation of c-Fos was almost completely blocked. The short splice form of FLIP, however, showed a rather stimulatory effect on c-Fos induction. Together these data demonstrate the existence of a death receptor-induced, FADD- and caspase-8-dependent pathway leading to c-Fos induction that is inhibited by the long splice form FLIP-L.
Resumo:
Fas(Apo-1/CD95), a receptor belonging to the tumor necrosis factor receptor family, induces apoptosis when triggered by Fas ligand. Upon its activation, the cytoplasmic domain of Fas binds several proteins which transmit the death signal. We used the yeast two-hybrid screen to isolate Fas-associated proteins. Here we report that the ubiquitin-conjugating enzyme UBC9 binds to Fas at the interface between the death domain and the membrane-proximal region of Fas. This interaction is also seen in vivo. UBC9 transiently expressed in HeLa cells bound to the co-expressed cytoplasmic segment of Fas. FAF1, a Fas-associated protein that potentiates apoptosis (Chu et al. (1996) Proc. Natl. Acad. Sci. USA 92, 11894-11898), was found to contain sequences similar to ubiquitin. These results suggest that proteins related to the ubiquitination pathway may modulate the Fas signaling pathway.
Resumo:
In ovarian follicles, cumulus cells provide the oocyte with small molecules that permit growth and control maturation. These nutrients reach the germinal cell through gap junction channels, which are present between the cumulus cells and the oocyte, and between the cumulus cells. In this study the involvement of intercellular communication mediated by gap junction channels on oocyte maturation of in vitro cultured bovine cumulus-oocyte complexes (COCs) was investigated. The stages of oocyte maturation were determined by Hoechst 33342 staining, which showed that 90% of COCs placed in the maturation medium for 24 h progress to the metaphase II stage. Bovine COC gap junction communication was disrupted initially using n-alkanols, which inhibit any passage through gap junctions. In the presence of 1-heptanol (3 mmol l(-1)) or octanol (3.0 mmol l(-1) and 0.3 mmol l(-1)), only 29% of the COCs reached metaphase II. Removal of the uncoupling agent was associated with restoration of oocyte maturation, indicating that treatment with n-alkanols was neither cytotoxic nor irreversible. Concentrations of connexin 43 (Cx43), the major gap junction protein expressed in the COCs, were decreased specifically using a recombinant adenovirus expressing the antisense Cx43 cDNA (Ad-asCx43). The efficacy of adenoviral infection was > 95% in cumulus cells evaluated after infection with recombinant adenoviruses expressing the green fluorescence protein. RT-PCR performed on total RNA isolated from Ad-asCx43-infected COCs showed that the rat Cx43 cDNA was transcribed. Western blot analysis revealed a three-fold decrease in Cx43 expression in COCs expressing the antisense RNA for Cx43. Injection of cumulus cells with Lucifer yellow demonstrated further that the resulting lower amount of Cx43 in infected COCs is associated with a two-fold decrease in the extent of coupling between cumulus cells. In addition, oocyte maturation was decreased by 50% in the infected COC cultures. These results indicate that Cx43-mediated communication between cumulus cells plays a crucial role in maturation of bovine oocytes.
Resumo:
Telomerase is a ribonucleoprotein complex responsible for the maintenance of the length of the telomeres during cell division, which is active in germ-line cells as well as in the vast majority of tumors but not in most normal tissues. The wide expression of the human telomerase catalytic subunit (hTERT) in tumors makes it an interesting candidate vaccine for cancer. hTERT-derived peptide 540-548 (hTERT(540)) has been recently shown to be recognized in an HLA-A*0201-restricted fashion by T cell lines derived from peptide-stimulated peripheral blood mononuclear cells (PBMC) from healthy donors. As a first step to the inclusion of this peptide in immunotherapy clinical trials, it is crucial to assess hTERT(540)-specific T cell reactivity in cancer patients as well as the ability of hTERT-specific CD8(+) T lymphocytes to recognize and lyse hTERT-expressing target cells. Here, we have analyzed the CD8(+) T cell response to peptide hTERT(540) in HLA-A*0201 melanoma patients by using fluorescent HLA-A*0201/hTERT(540) peptide tetramers. HLA-A*0201/hTERT(540) tetramer(+) CD8(+) T cells were readily detected in peptide-stimulated PBMC from a significant proportion of patients and could be isolated by tetramer-guided cell sorting. hTERT(540)-specific CD8(+) T cells were able to specifically recognize HLA-A*0201 cells either pulsed with peptide or transiently transfected with a minigene encoding the minimal epitope. In contrast, they failed to recognize hTERT-expressing HLA-A*0201(+) target cells. Furthermore, in vitro proteasome digestion studies revealed inadequate hTERT processing. Altogether, these results raise questions on the use of hTERT(540) peptide for cancer immunotherapy.
Resumo:
BACKGROUND: Up to now, the different uptake pathways and the subsequent intracellular trafficking of plasmid DNA have been largely explored. By contrast, the mode of internalization and the intracellular routing of an exogenous mRNA in transfected cells are poorly investigated and remain to be elucidated. The bioavailability of internalized mRNA depends on its intracellular routing and its potential accumulation in dynamic sorting sites for storage: stress granules and processing bodies. This question is of particular significance when a secure transposon-based system able to integrate a therapeutic transgene into the genome is used. Transposon vectors usually require two components: a plasmid DNA, carrying the gene of interest, and a source of transposase allowing the integration of the transgene. The principal drawback is the lasting presence of the transposase, which could remobilize the transgene once it has been inserted. Our study focused on the pharmacokinetics of the transposition process mediated by the piggyBac transposase mRNA transfection. Exogenous mRNA internalization and trafficking were investigated towards a better apprehension and fine control of the piggyBac transposase bioavailability. RESULTS: The mRNA prototype designed in this study provides a very narrow expression window of transposase, which allows high efficiency transposition with no cytotoxicity. Our data reveal that exogenous transposase mRNA enters cells by clathrin and caveolae-mediated endocytosis, before finishing in late endosomes 3 h after transfection. At this point, the mRNA is dissociated from its carrier and localized in stress granules, but not in cytoplasmic processing bodies. Some weaker signals have been observed in stress granules at 18 h and 48 h without causing prolonged production of the transposase. So, we designed an mRNA that is efficiently translated with a peak of transposase production 18 h post-transfection without additional release of the molecule. This confines the integration of the transgene in a very small time window. CONCLUSION: Our results shed light on processes of exogenous mRNA trafficking, which are crucial to estimate the mRNA bioavailability, and increase the biosafety of transgene integration mediated by transposition. This approach provides a new way for limiting the transgene copy in the genome and their remobilization by mRNA engineering and trafficking.
Resumo:
A defect in glucose sensing of the pancreatic beta-cells has been observed in several animal models of type II diabetes and has been correlated with a reduced gene expression of the glucose transporter type 2 (Glut2). In a transgenic mouse model, expression of Glut2 antisense RNA in pancreatic beta-cells has recently been shown to be associated with an impaired glucose-induced insulin secretion and the development of diabetes. To identify factors that may be involved in the specific decrease of Glut2 in the beta-cells of the diabetic animal, an attempt was made to localize the cis-elements and trans-acting factors involved in the control of Glut2 expression in the endocrine pancreas. It was demonstrated by transient transfection studies that only 338 base pairs (bp) of the murine Glut2 proximal promoter are needed for reporter gene expression in pancreatic islet-derived cell lines, whereas no activity was detected in nonpancreatic cells. Three cis-elements, GTI, GTII, and GTIII, have been identified by DNAse I footprinting and gel retardation experiments within these 338 bp. GTI and GTIII bind distinct but ubiquitously expressed trans-acting factors. On the other hand, nuclear proteins specifically expressed in pancreatic cell lines interact with GTII, and their relative abundance correlates with endogenous Glut2 expression. These GTII-binding factors correspond to nuclear proteins of 180 and 90 kilodaltons as defined by Southwestern analysis. The 180-kilodalton factor is present in pancreatic beta-cell lines but not in an alpha-cell line. Mutation of the GTI or GTIII cis-elements decreases transcriptional activity directed by the 338-bp promoter, whereas mutation of GTII increases gene transcription. Thus negative and positive regulatory sequences are identified within the proximal 338 bp of the GLUT2 promoter and may participate in the islet-specific expression of the gene by binding beta-cell specific trans-acting factors.
Resumo:
AIMS: Brugada syndrome (BrS) is characterized by arrhythmias leading to sudden cardiac death. BrS is caused, in part, by mutations in the SCN5A gene, which encodes the sodium channel alpha-subunit Na(v)1.5. Here, we aimed to characterize the biophysical properties and consequences of a novel BrS SCN5A mutation. METHODS AND RESULTS: SCN5A was screened for mutations in a male patient with type-1 BrS pattern ECG. Wild-type (WT) and mutant Na(v)1.5 channels were expressed in HEK293 cells. Sodium currents (I(Na)) were analysed using the whole-cell patch-clamp technique at 37 degrees C. The electrophysiological effects of the mutation were simulated using the Luo-Rudy model, into which the transient outward current (I(to)) was incorporated. A new mutation (C1850S) was identified in the Na(v)1.5 C-terminal domain. In HEK293 cells, mutant I(Na) density was decreased by 62% at -20 mV. Inactivation of mutant I(Na) was accelerated in a voltage-dependent manner and the steady-state inactivation curve was shifted by 11.6 mV towards negative potentials. No change was observed regarding activation characteristics. Altogether, these biophysical alterations decreased the availability of I(Na). In the simulations, the I(to) density necessary to precipitate repolarization differed minimally between the two genotypes. In contrast, the mutation greatly affected conduction across a structural heterogeneity and precipitated conduction block. CONCLUSION: Our data confirm that mutations of the C-terminal domain of Na(v)1.5 alter the inactivation of the channel and support the notion that conduction alterations may play a significant role in the pathogenesis of BrS.
Resumo:
Orphan receptors of the FTZ-F1-related group of nuclear receptors (xFF1r) were identified in Xenopus laevis by isolation of cDNAs from a neurula stage library. Two cDNAs were found, which encode full length, highly related receptor proteins, xFF1rA and B, whose closet relative known so far is the murine LRH-1 orphan receptor. xFF1rA protein expressed by a recombinant vaccinia virus system specifically binds to FTZ-F1 response elements (FRE; PyCAAGGPyCPu). In cotransfection studies, xFF1rA constitutively activates transcription, in a manner dependent on the number of FREs. The amounts of at least four mRNAs encoding full-length receptors greatly increase between gastrula and early tailbud stages and decrease at later stages. At early tailbud stages, xFTZ-F1-related antigens are found in all nuclei of the embryo.