62 resultados para Gene RECK metastasis suppressor

em Université de Lausanne, Switzerland


Relevância:

100.00% 100.00%

Publicador:

Resumo:

Metastases are responsible for most cancer-related deaths. One of the hallmarks of metastatic cells is increased motility and migration through extracellular matrixes. These processes rely on specific small GTPases, in particular those of the Rho family. Deleted in liver cancer-1 (DLC1) is a tumor suppressor that bears a RhoGAP activity. This protein is lost in most cancers, allowing malignant cells to proliferate and disseminate in a Rho-dependent manner. However, DLC1 is also a scaffold protein involved in alternative pathways leading to tumor and metastasis suppressor activities. Recently, substantial information has been gathered on these mechanisms and this review is aiming at describing the potential and known alternative GAP-independent mechanisms allowing DLC1 to impair migration, invasion, and metastasis formation.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Little is known about the mechanisms that establish the position of the division plane in eukaryotic cells. Wild-type fission yeast cells divide by forming a septum in the middle of the cell at the end of mitosis. Dmf1 mutants complete mitosis and initiate septum formation, but the septa that form are positioned at random locations and angles in the cell, rather than in the middle. We have cloned the dmf1 gene as a suppressor of the cdc7-24 mutant. The dmf1 mutant is allelic with mid1. The gene encodes a novel protein containing a putative nuclear localization signal, and a carboxy-terminal PH domain. In wild-type cells, Dmf1p is nuclear during interphase, and relocates to form a medial ring at the cell cortex coincident with the onset of mitosis. This relocalization occurs before formation of the actin ring and is associated with increased phosphorylation of Dmf1p. The Dmf1p ring can be formed in the absence of an actin ring, but depends on some of the genes required for actin ring formation. When the septum is completed and the cells separate, Dmf1p staining is once again nuclear. These data implicate Dmf1p as an important element in assuring correct placement of the division septum in Schizosaccharomyces pombe cells.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Cancer is the second leading cause of mortality worldwide. Cancer progression leads to metastasis formation, which accounts for more than ninety percent of cancer-related death. Metastases are more difficult to be surgically removed because of their invasive behavior and shape. In addition, during their transformation journey, they become more and more resistant to anticancer drugs. Significant improvements have been achieved in therapy against cancer in recent years but targeting the metastatic cascade remains the Achilles heel of the cure against cancer. A First step in the metastatic process is the escape of cancer cells from the primary tumor site. This involves an increase in cell motility and the concomitant ability to clear a path through the extracellular matrix. From a therapeutic point of view, inhibition of cell migration is a logical approach to develop anti-metastatic drugs. Our lab previously developed a cell permeable peptide derived from a caspase-3-generaied fragment of the RasGAP protein called TAT-RasGAP317-326. This peptide efficiently and specifically sensitizes cancer cells to chemotherapy- and radiotherapy-induced ceil death, which allows decreasing the anticancer drug doses and eventually their associated side- effects. In the present study we discovered that TAT-RasGAP317.326 also increases cell adhesion which was associated with inhibition of cell migration and invasion into the extracellular matrix. The ability of TAT-RasGAP317.326 to increase ceil adhesion involves the dramatic depolymerization of actin cytoskekton together with redistribution of focal adhesions. We found that the inhibitory effects on migration were mediated by a RhoGAP tumor and metastasis suppressor cailed DLC1 (Deleted in Liver Cancer 1). Moreover. DEC 1 was found to be a direct RasGAP-interacting protein and this interaction requires the RasGAP tryptophan 317 residue, the very first RasGAP residue of TAT-RasGAP317.326. We then evaluated the roie of RasGAP fragments in the in vivo metastatic cascade. We found that breast cancer cells overexpressing the parental RasGAP fragment, to which the TAT-RasGAP317.326 peptide belongs, have a markedly decreased ability to form lung metastases. Unfortunately, we were not able to recapitulate these an ti-metastatic effects when TAT-RasGAP317.326 was injected. However, we later understood that this was due to the fact that TAT-RasGAP317.326 was not properly delivered to the primary tumors. Further work, aimed at better understanding of how TAT-RasGAP317.326 functions, revealed that the ten amino acid TAT-RasGAP317.326 peptide could, be narrowed down to a three amino acid TAT-RasGAP317.329 peptide while keeping its sensitizer activity. In parallel, investigations on the RasGAP-DLCl binding indicated that the arginine linger of the DLC1 GAP domain is required for this interaction, which suggests that TAT-RasGAP317.326 modulates the GAP activity of DLC1. Additional work should be performed to fully elucidate its mechanism of action and render TAT-RasGAP317.326 usable as a tool to fight cancer on two fronts, by improving chemotherapy and preventing metastatic progression. - Le cancer est la deuxième cause de mortalité dans le monde. La formation de métastases est la dernière étape de la progression cancéreuse et représente plus du nonante pour cent des morts induites par le cancer. De par leur morphologie et comportement invasifs, ii est difficile d'avoir recours à la chirurgie pour exciser des métastases. De plus, les cellules cancéreuses en progression deviennent souvent de plus en plus résistantes aux drogues anticancéreuses. Ces dernières années, des avancements significatifs ont contribué à l'amélioration de la lutte contre le cancer. Néanmoins, pouvoir cibler spécifiquement la cascade métastatique demeure cependant le talon d'Achille des thérapies anticancéreuses. Une première étape dans ie processus métastatique est l'évasion des cellules cancéreuses du site de la tumeur primaire. Ceci requiert une augmentation de la motiliié cellulaire couplée à la capacité de se frayer un chemin au sein de la matrice extracelluiaire. D'un point de vue thérapeutique, inhiber la migration cellulaire est une approche attrayante. Notre laboratoire a développé un peptide, nommé TAT-RasGAP317.326 dérivé d'un fragment qui est lui-même le résultat du clivage de la protéine RasGAP par la caspase-3. Ce peptide est capable de pénétrer les cellules cancéreuses et de les sensibiliser spécifiquement à la mort induite par la radiothérapie et la chimiothérapie. La finalité des effets de ce peptide est de pouvoir diminuer les doses des traitements anti-cancéreux et donc des effets secondaires qu'ils engendrent. Dans cette étude, nous avons découvert que TAT-RasGAP317.326 augmente l'adhésion des cellules et inhibe la migration cellulaire ainsi que l'invasion des cellules à travers une matrice extracellulaire. La capacité de TAT-RasGAP317.326 à induire l'adhésion repose sur ia dépolymérisation du cytosquelette d'actine associée à une redistribution des points d'ancrage cellulaire. Nous avons découvert que l'inhibition de ia migration par TAT-RasGAP317.326 nécessitait la présence d'un suppresseur de tumeur et de métastases appelé DLC1 (Deleted in Liver Cancer l), qui par ailleurs s'avère aussi être une protéine RhoGAP. De plus, nous avons aussi trouvé que DLC1 était un partenaire d'interaction de RasGAP et que cette interaction s'effectuait via l'acide aminé tryptophane 317 de RasGAP. qui s'avère être le premier acide aminé du peptide TAT-RasGAP317.326. Nous avons ensuite évalué le rôle joué par certains fragments de RasGAP dans le processus de métastatisation. Dans ce contexte, des cellules de cancer du sein qui sur-expriment un fragment de RasGAP contenant la séquence TAT-RasGAP317.326 ont vu leur potentiel métastatique diminuer drastiquerment. Malheureusement, aucun effet anti-métastatique n'a été obtenu après injection de TAT-RasGAP317.326 dans les souris. Cependant, nous avons réalisé rétrospectivement que TAT-RasGAP317.326 n'était pas correctement délivré à la tumeur primaire, ce qui nous empêche de tirer des conclusions sur le rôle anti-métastatique de ce peptide. La suite de cette étude visant à mieux comprendre comment TAT-RasGAP317.326 agit, a mené à la découverte que les dix acides aminés de TAT-RasGAP317.326 pouvaient être réduits à trois acides aminés, TAT-RasGAP317.329, tout en gardant l'effet sensibilisateur à la chimiothérapie. En visant à élucider le mode d'interaction entre RasGAP et DLC1, nous avons découvert qu'un acide aminé nécessaire à l'activité GAP de DLC1 était requis pour lier RasGAP, ce qui laisse présager que TAT-RasGAp317.32c, module i'activité GAP de DLC1. Des travaux supplémentaires doivent encore être effectués pour complètement élucider les mécanismes d'action de TAT-RasGAP317.326 et afin de pouvoir l'utiliser comme un outil pour combattre le cancer sur deux fronts, en améliorant les chimiothérapies et en inhibant la formation de métastases.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

The peroxisome proliferator-activated receptor gamma (PPARgamma) plays a major role in fat tissue development and physiology. Mutations in the gene encoding this receptor have been associated to disorders in lipid metabolism. A thorough investigation of mice in which one PPARgamma allele has been mutated reveals that male PPARgamma heterozygous (PPARgamma +/-) mice exhibit a reduced body size associated with decreased body weight, reflecting lean mass reduction. This phenotype is reproduced when treating the mice with a PPARgamma- specific antagonist. Monosodium glutamate treatment, which induces weight gain and alters body growth in wild-type mice, further aggravates the growth defect of PPARgamma +/- mice. The levels of circulating GH and that of its downstream effector, IGF-I, are not altered in mutant mice. However, the IGF-I mRNA level is decreased in white adipose tissue (WAT) of PPARgamma +/- mice and is not changed by acute administration of recombinant human GH, suggesting an altered GH action in the mutant animals. Importantly, expression of the gene encoding the suppressor of cytokine signaling-2, which is an essential negative regulator of GH signaling, is strongly increased in the WAT of PPARgamma +/- mice. Although the relationship between the altered GH signaling in WAT and reduced body size remains unclear, our results suggest a novel role of PPARgamma in GH signaling, which might contribute to the metabolic disorder affecting insulin signaling in PPARgamma mutant mice.

Relevância:

50.00% 50.00%

Publicador:

Resumo:

Metastasis depends on the ability of tumor cells to establish a relationship with the newly seeded tissue that is conducive to their survival and proliferation. However, the factors that render tissues permissive for metastatic tumor growth have yet to be fully elucidated. Breast tumors arising during pregnancy display early metastatic proclivity, raising the possibility that pregnancy may constitute a physiological condition of permissiveness for tumor dissemination. Here we have shown that during murine gestation, metastasis is enhanced regardless of tumor type, and that decreased NK cell activity is responsible for the observed increase in experimental metastasis. Gene expression changes in pregnant mouse lung and liver were shown to be similar to those detected in premetastatic sites and indicative of myeloid cell infiltration. Indeed, myeloid-derived suppressor cells (MDSCs) accumulated in pregnant mice and exerted an inhibitory effect on NK cell activity, providing a candidate mechanism for the enhanced metastatic tumor growth observed in gestant mice. Although the functions of MDSCs are not yet understood in the context of pregnancy, our observations suggest that they may represent a shared mechanism of immune suppression occurring during gestation and tumor growth.

Relevância:

40.00% 40.00%

Publicador:

Resumo:

Cyclooxyganase-2 (COX-2), a rate-limiting enzyme in the prostaglandin synthesis pathway, is overexpressed in many cancers and contributes to cancer progression through tumor cell-autonomous and paracrine effects. Regular use of non-steroidal anti-inflammatory drugs or selective COX-2 inhibitors (COXIBs) reduces the risk of cancer development and progression, in particular of the colon. The COXIB celecoxib is approved for adjunct therapy in patients with Familial adenomatous polyposis at high risk for colorectal cancer (CRC) formation. Long-term use of COXIBs, however, is associated with potentially severe cardiovascular complications, which hampers their broader use as preventive anticancer agents. In an effort to better understand the tumor-suppressive mechanisms of COXIBs, we identified MAGUK with Inverted domain structure-1 (MAGI1), a scaffolding protein implicated in the stabilization of adherens junctions, as a gene upregulated by COXIB in CRC cells and acting as tumor suppressor. Overexpression of MAGI1 in CRC cell lines SW480 and HCT116 induced an epithelial-like morphology; stabilized E-cadherin and β-catenin localization at cell-cell junctions; enhanced actin stress fiber and focal adhesion formation; increased cell adhesion to matrix proteins and suppressed Wnt signaling, anchorage-independent growth, migration and invasion in vitro. Conversely, MAGI1 silencing decreased E-cadherin and β-catenin localization at cell-cell junctions; disrupted actin stress fiber and focal adhesion formation; and enhanced Wnt signaling, anchorage-independent growth, migration and invasion in vitro. MAGI1 overexpression suppressed SW480 and HCT116 subcutaneous primary tumor growth, attenuated primary tumor growth and spontaneous lung metastasis in an orthotopic model of CRC, and decreased the number and size of metastatic nodules in an experimental model of lung metastasis. Collectively, these results identify MAG1 as a COXIB-induced inhibitor of the Wnt/β-catenin signaling pathway, with tumor-suppressive and anti-metastatic activity in experimental colon cancer.

Relevância:

40.00% 40.00%

Publicador:

Resumo:

Chemotherapy is widely used as a systemic treatment modality in cancer patients and provides survival benefits for a significant fraction of treated patients H However, some patients suffer from cancer relapse and rapidly progress to metastasis, suggesting that following chemotherapy their residual tumor developed a more aggressive phenotype 4 5. Although some molecular mechanisms involved in chemo-resistance and chemotherapy-induced metastatic relapse have been reported, more investigations and understanding of these processes are necessary before any translation into the clinic might be considered. By using the syngeneic metastatic 4T1 murine breast cancer model, we observed that chemotherapy treatment and selection of chemotherapy-resistant cancer cells in vitro can induces two opposite phenotypes: a dormant one and a relapsing-metastatic one. Previous studies in our laboratory demonstrated that irradiation of mammary gland promotes tumor metastasis, at least in part, by inducing the recruitment of CD11b+ cells to both the primary tumor and the lungs at a pre-metastatic stage. In this study we found that CD11b+ cells may also play important roles in chemotherapy-induced tumor metastasis and dormancy in vivo. Tumor cells expressing the stem cell marker Sca-1 were enriched by chemotherapy treatment in vitro, as well as in tumor metastasis in vivo. Furthermore, tumor-derived CD11b+ cells were capable to maintain and expand this population in vitro. These results suggest that the expansion of a tumor cell population with stem cell features might be a mechanism by which chemotherapy induces metastasis. On the other hand, the same drug treatment in vitro generated resistant cells with a dormant phenotype. Dormant tumor cells were able to induce an in vivo immune- inflammatory response in the draining lymph node, which is normally absent due to the immunosuppressive effects of tumor-recruited myeloid derived- suppressor cells (MDSCs). Genome-wide gene expression analysis revealed the enrichment of invasion and metastasis-related genes in the relapsing metastatic tumor cells and immune response-related genes in the dormant tumor cells. Interestingly, CD11b+ cells derived from the microenvironment of growing-metastatic tumors, but not CD11b+ cells derived from the spleen of tumor-free mice, were able to instigate outgrowth of dormant tumor cells in vivo. Also, dormant cells formed growing and metastatic tumors when injected into immune-compromised NGS mice. These results point to a role of chemotherapy in enabling treated tumor cells to acquire immune response-inducing capabilities, while impairing the recruitment of CD11b+ cells and their differentiation into an immune-suppressive cell. The molecular mechanisms underneath these effects are being further investigated. In conclusion, results obtained in this model indicate that chemotherapy can induce a dormant phenotype in cancer cells and that this state of dormancy can be broken by MDSCs educated by relapsing tumors. Understanding the mechanism beyond these effects, in particular unraveling the genetic or epigenetic determinants of dormancy vs relapse, might open the way to therapies aimed and maintaining residual cells escaping chemotherapy in a state of sustained dormancy. - La chimiothérapie est un traitement systémique largement utilisé chez les patients cancéreux qui donne un avantage de survie significatif pour une bonne partie de patients traités (1-3). Cependant, certains patients souffrent d'une rechute et progressent ensuite vers la métastase. Ceci suggère que leur tumeur résiduelle a développé un phénotype agressif suite à la chimiothérapie (4-5). Bien que certains mécanismes moléculaires impliqués dans la chimiorésistance et la rechute métastatique ont été identifiés, d'avantage d'études sont nécessaires afin de mieux comprendre ce phénomène et de développer des nouvelles thérapies cliniques. En utilisant un modèle syngénique de cancer du sein métastatique chez la sourie (4T1), nous avons observé que la sélection des cellules cancéreuses résistantes à la chimiothérapie in vitro peut induire deux phénotypes opposés: un phénotype de dormance et un phénotype de progression métastatique. Une étude précédente issue de notre laboratoire a démontré que l'irradiation de la glande mammaire favorise la métastase de tumeurs recourants suite au recrutement de cellules CD11b+ dans la tumeur primaire et dans les poumons pré-métastatiques. Dans notre étude nous avons constaté que les cellules CD11b+ peuvent également jouer un rôle important dans la formation de métastases induites par la chimiothérapie ainsi que dans le maintien de la dormance in vivo. Nous avons également observé un enrichissement de cellules tumorales exprimant le marqueur de cellule souche Sca-1 parmi les cellules tumorales résistantes à la chimiothérapie et dans les cellules qui on formé des métastases in vivo. Des cellules CD11b+ dérivées du microenvironnement tumorale favorisent l'expansion de la population de cellules tumorales Sca-1+ in vitro. Ces résultats suggèrent que l'expansion d'une population de cellules tumorales avec des caractéristiques de cellules souches pourrait constituer un mécanisme par lequel la chimiothérapie induit des métastases dans des tumeurs récurrentes. D'autre part le même traitement de chimiothérapie peut générer des cellules résistantes avec un phénotype dormant. Les expériences in vivo indiquent que les cellules tumorales dormantes induisent une réponse immunitaire inflammatoire dans le ganglion lymphatique de drainage, qui est normalement réprimée par des cellules myéloïdes suppressives de tumeur (MDSC). Une analyse d'expression de gènes a révélé l'enrichissement de gènes liés à l'invasion et à la métastase dans les cellules tumorales récurrentes et des gènes liés à la réponse immunitaire dans les cellules tumorales dormantes. Les cellules CD11b+ issues du microenvironnement des tumeurs récurrents ont incité la croissance des cellules tumorales dormantes in vivo, tandis que les cellules CD11b+ dérivées de la rate de souris non porteuses de tumeur ne l'étaient pas. Les mécanismes moléculaires sous-jacents restent à découvrir. En conclusion, les résultats obtenus dans ce modèle indiquent que la chimiothérapie pourrait favoriser non seulement l'induction d'une dormance cellulaire, mais également que les cellules dormantes seraient adroits de induire une réponse immunitaire capable les maintenir dans un état de dormance prolongé. Un déséquilibre dans cette réponse immunitaire pourrait des lors briser cet état de dormance et induire une progression tumorale. Comprendre les mécanismes responsables de ces effets, en particulier l'identification des déterminants génétiques ou épigénétiques liés à la dormance vs la rechute, pourraient ouvrir la voie à des nouvelles thérapies visant le maintien d'un état de dormance permanente des cellules résiduelles après chimiothérapie.

Relevância:

40.00% 40.00%

Publicador:

Resumo:

Cheap and massively parallel methods to assess the DNA-binding specificity of transcription factors are actively sought, given their prominent regulatory role in cellular processes and diseases. Here we evaluated the use of protein-binding microarrays (PBM) to probe the association of the tumor suppressor AP2α with 6000 human genomic DNA regulatory sequences. We show that the PBM provides accurate relative binding affinities when compared to quantitative surface plasmon resonance assays. A PBM-based study of human healthy and breast tumor tissue extracts allowed the identification of previously unknown AP2α target genes and it revealed genes whose direct or indirect interactions with AP2α are affected in the diseased tissues. AP2α binding and regulation was confirmed experimentally in human carcinoma cells for novel target genes involved in tumor progression and resistance to chemotherapeutics, providing a molecular interpretation of AP2α role in cancer chemoresistance. Overall, we conclude that this approach provides quantitative and accurate assays of the specificity and activity of tumor suppressor and oncogenic proteins in clinical samples, interfacing genomic and proteomic assays.

Relevância:

40.00% 40.00%

Publicador:

Resumo:

Uveal melanoma is associated with a high mortality rate once metastases occur, with over >90% of metastatic patients dying within less than 1 year from metastases to the liver. The intraarterial hepatic (iah) administration of the alkylating agent fotemustine holds some promise with response rates of 36% and median survival of 15 months. Here, we investigated whether the DNA-repair-protein MGMT may be involved in the variability of response to fotemustine and temozolomide in uveal melanoma. Epigenetic inactivation of MGMT has been demonstrated to be a predictive marker for benefit from alkylating agent therapy in glioblastoma. We found a methylated MGMT promoter in 6% of liver metastases from 34 uveal melanoma patients. The mean MGMT activity measured in liver metastases with negligible liver tissue content was significantly lower than in liver tissue (146 versus 523 fmol/mg protein, p = 0.002). Expression of the MGMT protein was detectable in 50% of 88 metastases by immunohistochemistry on a tissue microarray. Expression was heterogeneous, and in accordance with MGMT activity data, usually lower than in the surrounding liver. Differential MGMT activity/expression between metastasis and liver tissue and more efficient depletion of MGMT with higher doses of alkylating agent therapy using iah delivery may provide the pharmacologic window for the higher response rate. However, these results do not support MGMT methylation status or protein expression as predictive markers for treatment outcome to iah chemotherapy with alkylating agents.

Relevância:

40.00% 40.00%

Publicador:

Resumo:

Cancer progression is dependent, in part, on interactions between tumor cells and the host microenvironment. During pregnancy, physiological changes occur that include inflammation and reduced immunity, both of which can promote tumor growth. Accordingly, tumors are observed to be more aggressive and to have greater proclivity toward metastasis during pregnancy. In this work, myeloid-derived suppressor cells (MDSC), a population of heterogeneous and pluripotent cells that can down-regulate immune responses during pathological conditions, were studied in the context of mouse and human gestation. The gene expression profile of mouse MDSC has been shown to differ in pregnant and virgin mice, and the profile in pregnant animals bears similarity to that of MDSC associated with the tumor microenvironment. Common induced genes include Fibronectin1 and Olfactomedin4, which are known to be involved in extracellular matrix remodeling and tissue permissiveness to tumor cells implantation. Our observations suggest that mouse MDSC may represent a shared regulatory mechanism of tissue permissiveness that occurs during the physiological state of gestation and tumor growth. Pregnancy-associated changes in immunosuppressive myeloid cell activity have also been studied in humans. We show that CD33+ myeloid cells isolated from PBMC (peripheral blood mononuclear cells) of pregnant women are more strongly immunosuppressive on T cells than CD33+ cells removed from non-pregnant subjects. During murine gestation, decreased natural killer (NK) cell activity is responsible, at least in part, for the increase in experimental metastasis. However, although peripheral blood NK cell numbers and cytotoxicity were slightly reduced in pregnant women, neither appeared to be regulated by CD33+ cells. Nevertheless, based on its observed suppression of T cell responses, the CD33+ PBMC subset appears to be an appropriate myeloid cell population to study in order to elucidate mechanisms of immune regulation that occur during human pregnancy. Our findings regarding the immunosuppressive function of CD33+cells and the role of NK cells during human pregnancy are consistent with the notion that changes in the function of the immune system participate in the constitution of a permissive soil for tumour progression.

Relevância:

40.00% 40.00%

Publicador:

Resumo:

Background and aim of the study: Genomic gains and losses play a crucial role in the development and progression of DLBCL and are closely related to gene expression profiles (GEP), including the germinal center B-cell like (GCB) and activated B-cell like (ABC) cell of origin (COO) molecular signatures. To identify new oncogenes or tumor suppressor genes (TSG) involved in DLBCL pathogenesis and to determine their prognostic values, an integrated analysis of high-resolution gene expression and copy number profiling was performed. Patients and methods: Two hundred and eight adult patients with de novo CD20+ DLBCL enrolled in the prospective multicentric randomized LNH-03 GELA trials (LNH03-1B, -2B, -3B, 39B, -5B, -6B, -7B) with available frozen tumour samples, centralized reviewing and adequate DNA/RNA quality were selected. 116 patients were treated by Rituximab(R)-CHOP/R-miniCHOP and 92 patients were treated by the high dose (R)-ACVBP regimen dedicated to patients younger than 60 years (y) in frontline. Tumour samples were simultaneously analysed by high resolution comparative genomic hybridization (CGH, Agilent, 144K) and gene expression arrays (Affymetrix, U133+2). Minimal common regions (MCR), as defined by segments that affect the same chromosomal region in different cases, were delineated. Gene expression and MCR data sets were merged using Gene expression and dosage integrator algorithm (GEDI, Lenz et al. PNAS 2008) to identify new potential driver genes. Results: A total of 1363 recurrent (defined by a penetrance > 5%) MCRs within the DLBCL data set, ranging in size from 386 bp, affecting a single gene, to more than 24 Mb were identified by CGH. Of these MCRs, 756 (55%) showed a significant association with gene expression: 396 (59%) gains, 354 (52%) single-copy deletions, and 6 (67%) homozygous deletions. By this integrated approach, in addition to previously reported genes (CDKN2A/2B, PTEN, DLEU2, TNFAIP3, B2M, CD58, TNFRSF14, FOXP1, REL...), several genes targeted by gene copy abnormalities with a dosage effect and potential physiopathological impact were identified, including genes with TSG activity involved in cell cycle (HACE1, CDKN2C) immune response (CD68, CD177, CD70, TNFSF9, IRAK2), DNA integrity (XRCC2, BRCA1, NCOR1, NF1, FHIT) or oncogenic functions (CD79b, PTPRT, MALT1, AUTS2, MCL1, PTTG1...) with distinct distribution according to COO signature. The CDKN2A/2B tumor suppressor locus (9p21) was deleted homozygously in 27% of cases and hemizygously in 9% of cases. Biallelic loss was observed in 49% of ABC DLBCL and in 10% of GCB DLBCL. This deletion was strongly correlated to age and associated to a limited number of additional genetic abnormalities including trisomy 3, 18 and short gains/losses of Chr. 1, 2, 19 regions (FDR < 0.01), allowing to identify genes that may have synergistic effects with CDKN2A/2B inactivation. With a median follow-up of 42.9 months, only CDKN2A/2B biallelic deletion strongly correlates (FDR p.value < 0.01) to a poor outcome in the entire cohort (4y PFS = 44% [32-61] respectively vs. 74% [66-82] for patients in germline configuration; 4y OS = 53% [39-72] vs 83% [76-90]). In a Cox proportional hazard prediction of the PFS, CDKN2A/2B deletion remains predictive (HR = 1.9 [1.1-3.2], p = 0.02) when combined with IPI (HR = 2.4 [1.4-4.1], p = 0.001) and GCB status (HR = 1.3 [0.8-2.3], p = 0.31). This difference remains predictive in the subgroup of patients treated by R-CHOP (4y PFS = 43% [29-63] vs. 66% [55-78], p=0.02), in patients treated by R-ACVBP (4y PFS = 49% [28-84] vs. 83% [74-92], p=0.003), and in GCB (4y PFS = 50% [27-93] vs. 81% [73-90], p=0.02), or ABC/unclassified (5y PFS = 42% [28-61] vs. 67% [55-82] p = 0.009) molecular subtypes (Figure 1). Conclusion: We report for the first time an integrated genetic analysis of a large cohort of DLBCL patients included in a prospective multicentric clinical trial program allowing identifying new potential driver genes with pathogenic impact. However CDKN2A/2B deletion constitutes the strongest and unique prognostic factor of chemoresistance to R-CHOP, regardless the COO signature, which is not overcome by a more intensified immunochemotherapy. Patients displaying this frequent genomic abnormality warrant new and dedicated therapeutic approaches.

Relevância:

40.00% 40.00%

Publicador:

Resumo:

Abstract : The Notch pathway is an important regulator of differentiation and carcinogenesis. In keratinocytes and possibly other specific epithelial cell types, it acts as tumour suppressor. Expression of endogenous Notch1 gene is markedly reduced in keratinocyte-derived squamous cell carcinoma (SCC) and cervical cancer cells, as well as in prostate cancer cell lines, and this difference is, at least in part, at the transcriptional level. Little is known on transcriptional control of the Notch1 gene with the exception that it is a p53-target. Our work focused on the mechanisms involved in the different transcription level of the Notch1 gene in normal versus cancer cells. We show that the fully active minimal Notch1 promoter is differentially controlled in normal versus cancer cells. It consists of two distinct regions, one downstream of the transcription start site, which is likely to bind the basic transcription apparatus, and one upstream region characterized by highly GC-rich sequence. This latter region binds Sp/KLF family members, specifically Spa and KLF4, which is upregulated in cancer cells. This is functionally significant as KLF4 overexpression is sufficient to downmodulate Notchl gene transcription, while KLF4 knockdown, in combination with Spa, results in Notch1 upregulation. Control of Notch1 by KLF4/Sp3 is independent of p53. Biochemically, KLF4/Sp3 seem to affect preferentially the initiation step of Notch1 gene transcription, while p53 controls both initiation and elongation steps. Thus, the Notch1 gene is a negative Sp3/KLF4-target and this mechanism contributes, in parallel with p53, to Notch1 downregulation in cancer. Résumé : La voie de signalisation induite par Notch est considérablement impliquée dans la différenciation des cellules et dans la carcinogénèse. Dans les kératinocytes ainsi que dans d'autres types cellulaires de l'épithelium, il agit comme suppresseur de tumeur. L'expression endogène de Notch1 est remarquablement réduite dans les cellules du carcinome spino-cellulaire et du cancer du col de l'utérus ou dans les lignées cellulaires du cancer de la prostate. Cette différence s'explique, du moins en partie, par le niveau de transcription. Peu de choses sont connues sur le contrôle transcriptionnel de Notch1 à l'exception du fait qu'il soit une cible de p53. Notre travail s'est concentré sur les mécanismes impliqués dans la transcription de Notch1, mécanismes qui diffèrent entre les cellules normales et les cellules cancéreuses. Nous avons trouvé la plus petite région du promoteur de Notch1 qui est suffisante pour induire un haut niveau transcriptionnel et qui est contrôlée différemment dans les cellules normales et les cellules cancéreuses. Elle est constituée de deux régions distinctes: une en aval du site de départ de la transcription, qui lie probablement le complexe de base pour la transcription, et une en amont caractérisée par une séquence riche en GC. Cette région lie les membres de la famille Sp/KLF, spécifiquement Sp3 et KLF4, qui sont surexprimés dans les cellules cancéreuses. Ceci est fonctionnellement significatif car la surexpression de KLF4 dans les kératinocytes est suffisante pour diminuer la transcription de Notch1, alors que l'inhibition de KLF4 et de Spa, résulte en une augmentation de Notch1. En outre, le contrôle de Notch1 par KLF4 et Spa est indépendant de p53. Biochimiquement, KLF4 et Spa semblent plutôt affecter l'initiation de la transcription de Notch1 alors que p53 contrôle aussi bien l'initiation que l'élongation. En conclusion, le gène Notch1 est inhibé par Spa et KLF4: ce mécanisme contribue, en parallèle à p53, à diminuer l'expression de Notch1 dans les cellules cancéreuses.

Relevância:

40.00% 40.00%

Publicador:

Resumo:

THESIS SUMMARY : Metastasis is a multistep process involving tumour cell-autonomous features, the host tissue stroma of the primary tumour, the blood or lymphatic system as well as a receptive target organ. Most studies on factors influencing metastasis have concentrated on the characteristics of the disseminating tumour cell and on early steps of metastasis including invasion and angiogenesis. Although these steps are necessary for tumour cells to disseminate, it is the challenges encountered in the later steps of metastasis -survival while in the circulation and engraftment and outgrowth in the target organ -that account for the inefficiency of circulating tumour cells in establishing secondary lesions. Full understanding of the metastatic process therefore requires elucidation of the mechanisms that regulate these late steps, and in particular that determine what makes any given tissue permissive for metastatic tumour growth. To address this issue, we assessed the mechanisms whereby a physiological situation -pregnancy -can alter host permissiveness toward metastasis. We show that pregnant NOD/SCID mice -injected intravenously with tumour cells -develop more metastases than their non-pregnant counterparts irrespective of the tumour cell type. There was no direct effect of pregnancy-related circulating factors on tumour cell proliferation, and subcutaneous tumour growth does not vary between pregnant and nonpregnant animals. However, decreased elimination of tumour cells from the lung microvasculature was observed in pregnant mice, prompting us to assess whether pregnancy-related adaptations in innate immunity could account for this differential clearing. We found that natural killer (NK) cell fractions are decreased in blood and spleen of pregnant mice and that NK cell cytotoxicity is impaired, as reported previously. The use of NK-deficient mice or tumour cell lines resistant to NK killing abrogates the difference in metastasis load between pregnant and virgin mice. CD11 b+ Gr-1+ myeloid-derived suppressor cells (MDSC) have previously been shown to accumulate in tumour-bearing mice and to down-modulate NK activity. Accordingly, we show an increase in MDSC in pregnant mouse blood, spleen, lungs and liver. Depletion of MDSC prior to tumour cell injection decreased metastasis load in pregnant NOD/SCID mice but had no effect on virgin mice. Similarly, adoptive transfer of MDSC extracted from pregnant mice into virgin mice lead to increased metastasis take. In parallel, we investigated whether the lung and liver microenvironments are modified during pregnancy thereby providing a more "permissive soil" for the establishment of metastases. A comparative analysis of microarray data of pregnant mouse lungs and liver with "premetastatic niche" gene expression profiles of these organs shows that similar mechanisms could mediate an increase in lung and liver metastasis in pregnant mice and in mice harbouring an aggressive primary tumour. Several commonly up-regulated genes point towards the recruitment of myeloid cells, consistent with the accumulation of MDSC observed in pregnant mice. MDSC have never been evoked in the context of pregnancy before. Although the role of MDSC in pregnancy requires further investigation we suggest that MDSC accumulation constitutes an important and hitherto unrecognised common denominator of maternal immune tolerance and cancer immune escape. RESUME DE THESE : La métastatisation est un processus en plusieurs étapes qui implique des compétences particulières chez les cellules tumorales, le stroma de la tumeur primaire, les vaisseaux sanguins ou lymphatiques ainsi qu'un organe cible' réceptif. Jusqu'alors, la recherche s'est principalement intéressée aux facteurs qui influencent les étapes précoces de la métastatisation donc aux caractéristiques de la cellule métastatique, et aux processus tels que l'invasion et l'angiogenèse, tandis que peu d'études traitent des étapes tardives tel que la survie dans la circulation sanguine et l'établissement d'une lésion dans l'organe cible. En particulier, l'élucidation des facteurs qui déterminent la permissivité d'un tissu à la greffe de cellules disséminantes est indispensable à la compréhension de ce processus complexe qu'est la métastatisation. Nous proposons ici un modèle de souris récapitulant les étapes tardives de la métastatisation dans un contexte d'une permissivité accrue aux métastases chez la souris gravide, et nous évaluons les mécanismes impliqués. Les souris gestantes développent plus de métastases après l'injection intraveineuse de cellules tumorales, indépendamment du type de tumeur d'origine. Les taux élevés d'hormones et de facteurs de croissance chez la souris gravide n'inflúencent pas la prolifération des cellules tumorales et fa croissance de tumeurs sous-cutanées n'est pas non plus accélérée par la gestation. En revanche, une fois injectées, les cellules tumorales sont éliminées ` moins rapidement des vaisseaux pulmonaires chez la souris gravide que chez les contrôles. Cette observation est compatible avec un effet de la gestation sur l'immunité innée et nous avons mis en évidence une diminution des proportions de cellules NK (natural killer) dans le sang et la rate en particulier, ainsi qu'une cytotoxicité moindre envers des cellules tumorales. En utilisant des souris déficientes en cellules NK ou en injectant des cellules résistantes à l'attaqué par des cellules NK, la différence entre souris gestantes et non-gestantes disparaît. Il a été démontré chez des souris porteuses de tumeurs, que l'accumulation de cellules immunosuppressives de la lignée myélo-monocytaire (ou MDSC pour myeloid-derived suppressor tells) pouvait être responsable d'une inhibition de l'activité de cellules NK. Des nombres augmentés de ces cellules, caractérisées par les marqueurs de surface CD11b et Gr-1, ont été trouvés dans le sang, la rate, les poumons et le foie de souris gravides. Leur rôle dans la métastatisation est démontré par le fait que leur dépletion diminue le nombre de lésions secondaires chez la souris gestante, tandis que leur transfert dans des souris non-gestantes augmente le taux de métastases. L'utilisation de puces à ADN sur les foies et poumons de souris gravides a permis de mettre en évidence des différences d'expression génique proches de celles observées dans l'établissement de niches pré-métastatiques. Ceci suggère que des mécanismes similaires pourraient être responsables d'une permissivité accrue aux métastases chez la souris gravide et chez la souris porteuse d'une tumeur primaire agressive, telle que, en particulier, l'accumulation de cellules immunosuppressives dans les organes cibles. C'est la première fois que l'accumulation de MDSC est évoquée chez la souris gravide et nous proposons ici que celles-ci jouent un rôle dans la tolérance immunitaire envers le foetus et sont responsables de l'échappement de cellules tumorales injectées à la surveillance immunitaire par des cellules NK.

Relevância:

40.00% 40.00%

Publicador:

Resumo:

Natural killer cell lymphoma (NKCL) constitutes a rare and aggressive form of non-Hodgkin lymphoma, and there is little insight into its pathogenesis. Here we show that PRDM1 is a tumor suppressor gene in NKCLs that is inactivated by a combination of monoallelic deletion and promoter CpG island hypermethylation. We observed monoallelic deletion of PRDM1 loci in 8 of 18 (44%) NKCL cases. The other allele showed significant promoter methylation in 12 of 17 (71%) cases. In support of its role as a tumor suppressor gene, the reconstitution of PRDM1 in PRDM1-null NK cell lines led to G2/M cell cycle arrest, increased apoptosis, and a strong negative selection pressure with progressive elimination of PRDM1-expressing cells, which was enhanced when IL-2 concentration is limiting. We observed a progressive increase in PRDM1 expression-in particular, PRDM1α-in normal NK cells in response to IL-2 and in normal NK cells activated with an engineered NK cell target, K562-Cl9-mb21, suggesting its role in NK cell homeostasis. In support of this role, knockdown of PRDM1 by shRNA in normal NK cells resulted in the positive selection of these cells. We identified MYC and 4-1BBL as targets of PRDM1 in NK cells. Disruption of homeostatic control by PRDM1 may be an important pathogenetic mechanism for NKCL.

Relevância:

40.00% 40.00%

Publicador:

Resumo:

Exposing the human bronchial epithelial cell line BEAS-2B to the nitric oxide (NO) donor sodium 1-(N,N-diethylamino)diazen-1-ium-1, 2-diolate (DEA/NO) at an initial concentration of 0.6 mM while generating superoxide ion at the rate of 1 microM/min with the hypoxanthine/xanthine oxidase (HX/XO) system induced C:G-->T:A transition mutations in codon 248 of the p53 gene. This pattern of mutagenicity was not seen by 'fish-restriction fragment length polymorphism/polymerase chain reaction' (fish-RFLP/PCR) on exposure to DEA/NO alone, however, exposure to HX/XO led to various mutations, suggesting that co-generation of NO and superoxide was responsible for inducing the observed point mutation. DEA/NO potentiated the ability of HX/XO to induce lipid peroxidation as well as DNA single- and double-strand breaks under these conditions, while 0.6 mM DEA/NO in the absence of HX/XO had no significant effect on these parameters. The results show that a point mutation seen at high frequency in certain common human tumors can be induced by simultaneous exposure to reactive oxygen species and a NO source.