8 research outputs found

    ASPM-associated stem cell proliferation is involved in malignant progression of gliomas and constitutes an attractive therapeutic target

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>ASPM (<it>Abnormal Spindle-like Microcephaly associated</it>) over-expression was recently implicated in the development of malignant gliomas.</p> <p>Results</p> <p>To better characterize the involvement of ASPM in gliomas, we investigated the mRNA expression in 175 samples, including 8 WHO Grade II, 75 WHO Grade III and 92 WHO Grade IV tumors. <it>Aspm </it>expression was strongly correlated with tumor grade and increased at recurrence when compared to the initial lesion, whatever the initial grade of the primary tumor. ASPM expression also increased over serial passages in gliomaspheres <it>in vitro </it>and in mouse xenografts <it>in vivo</it>. Lentivirus-mediated shRNA silencing of ASPM resulted in dramatic proliferation arrest and cell death in two different gliomasphere models.</p> <p>Conclusion</p> <p>These data suggest that ASPM is involved in the malignant progression of gliomas, possibly through expansion of a cancer stem cell compartment, and is an attractive therapeutic target in glioblastoma multiforme.</p

    Study by interference RNA of aspm gene expression in tumor stem cells of high grade glioma

    No full text
    Les gliomes sont les tumeurs cĂ©rĂ©brales primitives les plus frĂ©quentes de l’adulte. Le glioblastome (grade IV) en est la forme la plus agressive, caractĂ©risĂ© par sa rĂ©sistance aux traitements actuels (chirurgie, chimiothĂ©rapie et radiothĂ©rapie). La mortalitĂ© de cette pathologie est quasi constante (survie mĂ©diane de 15 mois), ce qui justifie l’importance de dĂ©couvrir de nouvelles cibles thĂ©rapeutiques. Le challenge est d'arriver Ă  identifier des marqueurs spĂ©cifiques pour proposer un schĂ©ma thĂ©rapeutique alignant des stratĂ©gies de thĂ©rapies ciblĂ©es qui vont amĂ©liorer la prise en charge clinique, la survie globale et la survie sans progression des patients atteints de ces pathologies. Deux axes sont au centre des recherches fondamentales, translationnelles et cliniques. Le premier axe se dĂ©finit autour du dĂ©veloppement de molĂ©cules inhibitrices des voies de signalisation et le second autour du concept de cellules souches tumorales (CST) de glioblastomes (GBM) dĂ©couvertes rĂ©cemment dans le cerveau et qui rĂ©volutionnent la conception de la transformation tumorale.ASPM (Abnormal Spindle Like MicrocĂ©phaly Associated) est une cible candidate pertinente susceptible de participer au dĂ©veloppement des gliomes (Horvath et al., 2007 ; Hagmann et al., 2008). Cette protĂ©ine rĂ©gule la prolifĂ©ration des neuroblastes, elle est fortement exprimĂ©e au stade embryonnaire, mais, reste faiblement exprimĂ©e dans le cerveau adulte. Par ailleurs, ASPM est impliquĂ©e dans divers processus de cancĂ©risation (surexprimĂ©e dans les cancers du sein, du foie et du cerveau
), toute fois, le mĂ©canisme responsable de cette dĂ©rĂ©gulation n’est pas encore bien caractĂ©risĂ©.Nos Ă©tudes menĂ©es sur une sĂ©rie de 169 gliomes humains, sĂ©lectionnĂ©s Ă  partir de notre cohorte de patients, montrent que le gĂšne ASPM est un marqueur de la progression vers la malignitĂ©, les grades les plus Ă©levĂ©s exprimant le plus fortement ASPM. En outre, nous avons Ă©galement montrĂ© que le niveau des transcrits d’ASPM est augmentĂ© dans les rĂ©cidives de gliomes et qu’en in vitro, ASPM contrĂŽle la formation des gliomasphĂšres (CST de GBM) avec une augmentation de l’expression de ses transcrits dans les cultures in vitro au fil des passages. En continuitĂ© de ces observations, nous avons alors dĂ©veloppĂ© un sh-miR-RNA spĂ©cifique d’ASPM permettant l’extinction post-transcriptionnelle de ce gĂšne. Les rĂ©sultats obtenus in vitro montrent que la perte d’expression d’ASPM conduit Ă  un arrĂȘt de la prolifĂ©ration et aboutit Ă  une mort cellulaire massive.Actuellement, des modĂšles de greffe de gliomasphĂšres chez la souris (orthotopique) sont en cours de dĂ©veloppement pour confirmer les effets observĂ©s in vitro et vĂ©rifier in vivo la validitĂ© de notre approche thĂ©rapeutique. En perspective, nous tenterons d’étudier les effets du silencing d’ASPM sur la voie de signalisation la plus dĂ©rĂ©gulĂ©e (pRB / E2F ou PI3K / AKT). Enfin, nous Ă©tudierons le rĂŽle potentiel de cette protĂ©ine dans le contrĂŽle du cycle cellulaire, et, in fine la mise en Ă©vidence de ses partenaires
Glioblastoma (GBM) is the most frequent and aggressive form of primary brain tumors in adults; it is characterized by its resistance to current treatments (surgery, chemotherapy and radiotherapy). The prognosis is grim with a median survival of only 15 months underlining the importance to develop new therapeutic strategies. The recent development of the “tumor stem cell” (TSC) concept in hemopathies has been secondarily applied to gliomas with the identification of subpopulations of GBM cells which express neural stem cell markers and fulfill the criteria for stemness. Some evidences also suggest that this subpopulation could play a primary role in resistance to radio- and chemotherapy.ASPM (Abnormal Spindle Like Microcephaly Associated) is a protein regulating the proliferation of neuroblasts, highly expressed in the embryonic stage but weakly expressed in the adult brain. Preliminary reports suggesting that it could be involved in the development of gliomas (Horvath et al., 2007, Hagemann et al., 2008) prompted us to analyze further the role of this protein, focusing on its potential as a relevant candidate therapeutic target. In a series of 175 gliomas samples of various grades, we found that ASPM mRNA expression was strongly correlated with increasing tumor grade. We also found that ASPM expression increased at recurrence when compared to the initial lesion. Subsequently, we could demonstrate in vitro and in vivo that ASPM expression also increased over serial passages in gliomaspheres and in a mouse glioma xenograft model. In a therapeutic perspective, the effect of lentivirus-mediated shRNA post-transcriptional silencing of ASPM was evaluated in two different gliomasphere models and a dramatic proliferation arrest and cell death was observed. Taken together, these data suggest that ASPM is involved in the malignant progression of gliomas, possibly through expansion of a cancer stem cell compartment, and could be an attractive therapeutic target in glioblastoma multiforme.Another potential candidate tumor stem cell target in glioma is the sonic hedgehog pathway (hedgehog-Gli) which is required for GBM growth and stem cell expansion. In a collaborative study, it was found that NANOG, a transcription factor critically involved with self-renewal of undifferentiated embryonic stem cells, modulates gliomasphere clonogenicity, CD133+ stem cell behavior and proliferation. NANOG was regulated by hedgehog-Gli signalling and was essential for GBM tumourigenicity in orthotopic xenografts suggesting that it could also be a useful potential therapeutic target.Conclusions: Accumulating evidences suggest that tumor stem cells play an important role in the oncogenesis of gliomas and in their resistance to treatment. Our data support this concept and suggest that specific stemness markers may become useful targets to improve treatment of this devastating disease

    Etude par ARN interfĂ©rence de l’expression du gĂšne ASPM dans les cellules souches tumorales des gliomes de haut grade

    No full text
    Glioblastoma (GBM) is the most frequent and aggressive form of primary brain tumors in adults; it is characterized by its resistance to current treatments (surgery, chemotherapy and radiotherapy). The prognosis is grim with a median survival of only 15 months underlining the importance to develop new therapeutic strategies. The recent development of the “tumor stem cell” (TSC) concept in hemopathies has been secondarily applied to gliomas with the identification of subpopulations of GBM cells which express neural stem cell markers and fulfill the criteria for stemness. Some evidences also suggest that this subpopulation could play a primary role in resistance to radio- and chemotherapy.ASPM (Abnormal Spindle Like Microcephaly Associated) is a protein regulating the proliferation of neuroblasts, highly expressed in the embryonic stage but weakly expressed in the adult brain. Preliminary reports suggesting that it could be involved in the development of gliomas (Horvath et al., 2007, Hagemann et al., 2008) prompted us to analyze further the role of this protein, focusing on its potential as a relevant candidate therapeutic target. In a series of 175 gliomas samples of various grades, we found that ASPM mRNA expression was strongly correlated with increasing tumor grade. We also found that ASPM expression increased at recurrence when compared to the initial lesion. Subsequently, we could demonstrate in vitro and in vivo that ASPM expression also increased over serial passages in gliomaspheres and in a mouse glioma xenograft model. In a therapeutic perspective, the effect of lentivirus-mediated shRNA post-transcriptional silencing of ASPM was evaluated in two different gliomasphere models and a dramatic proliferation arrest and cell death was observed. Taken together, these data suggest that ASPM is involved in the malignant progression of gliomas, possibly through expansion of a cancer stem cell compartment, and could be an attractive therapeutic target in glioblastoma multiforme.Another potential candidate tumor stem cell target in glioma is the sonic hedgehog pathway (hedgehog-Gli) which is required for GBM growth and stem cell expansion. In a collaborative study, it was found that NANOG, a transcription factor critically involved with self-renewal of undifferentiated embryonic stem cells, modulates gliomasphere clonogenicity, CD133+ stem cell behavior and proliferation. NANOG was regulated by hedgehog-Gli signalling and was essential for GBM tumourigenicity in orthotopic xenografts suggesting that it could also be a useful potential therapeutic target.Conclusions: Accumulating evidences suggest that tumor stem cells play an important role in the oncogenesis of gliomas and in their resistance to treatment. Our data support this concept and suggest that specific stemness markers may become useful targets to improve treatment of this devastating disease.Les gliomes sont les tumeurs cĂ©rĂ©brales primitives les plus frĂ©quentes de l’adulte. Le glioblastome (grade IV) en est la forme la plus agressive, caractĂ©risĂ© par sa rĂ©sistance aux traitements actuels (chirurgie, chimiothĂ©rapie et radiothĂ©rapie). La mortalitĂ© de cette pathologie est quasi constante (survie mĂ©diane de 15 mois), ce qui justifie l’importance de dĂ©couvrir de nouvelles cibles thĂ©rapeutiques. Le challenge est d'arriver Ă  identifier des marqueurs spĂ©cifiques pour proposer un schĂ©ma thĂ©rapeutique alignant des stratĂ©gies de thĂ©rapies ciblĂ©es qui vont amĂ©liorer la prise en charge clinique, la survie globale et la survie sans progression des patients atteints de ces pathologies. Deux axes sont au centre des recherches fondamentales, translationnelles et cliniques. Le premier axe se dĂ©finit autour du dĂ©veloppement de molĂ©cules inhibitrices des voies de signalisation et le second autour du concept de cellules souches tumorales (CST) de glioblastomes (GBM) dĂ©couvertes rĂ©cemment dans le cerveau et qui rĂ©volutionnent la conception de la transformation tumorale.ASPM (Abnormal Spindle Like MicrocĂ©phaly Associated) est une cible candidate pertinente susceptible de participer au dĂ©veloppement des gliomes (Horvath et al., 2007 ; Hagmann et al., 2008). Cette protĂ©ine rĂ©gule la prolifĂ©ration des neuroblastes, elle est fortement exprimĂ©e au stade embryonnaire, mais, reste faiblement exprimĂ©e dans le cerveau adulte. Par ailleurs, ASPM est impliquĂ©e dans divers processus de cancĂ©risation (surexprimĂ©e dans les cancers du sein, du foie et du cerveau
), toute fois, le mĂ©canisme responsable de cette dĂ©rĂ©gulation n’est pas encore bien caractĂ©risĂ©.Nos Ă©tudes menĂ©es sur une sĂ©rie de 169 gliomes humains, sĂ©lectionnĂ©s Ă  partir de notre cohorte de patients, montrent que le gĂšne ASPM est un marqueur de la progression vers la malignitĂ©, les grades les plus Ă©levĂ©s exprimant le plus fortement ASPM. En outre, nous avons Ă©galement montrĂ© que le niveau des transcrits d’ASPM est augmentĂ© dans les rĂ©cidives de gliomes et qu’en in vitro, ASPM contrĂŽle la formation des gliomasphĂšres (CST de GBM) avec une augmentation de l’expression de ses transcrits dans les cultures in vitro au fil des passages. En continuitĂ© de ces observations, nous avons alors dĂ©veloppĂ© un sh-miR-RNA spĂ©cifique d’ASPM permettant l’extinction post-transcriptionnelle de ce gĂšne. Les rĂ©sultats obtenus in vitro montrent que la perte d’expression d’ASPM conduit Ă  un arrĂȘt de la prolifĂ©ration et aboutit Ă  une mort cellulaire massive.Actuellement, des modĂšles de greffe de gliomasphĂšres chez la souris (orthotopique) sont en cours de dĂ©veloppement pour confirmer les effets observĂ©s in vitro et vĂ©rifier in vivo la validitĂ© de notre approche thĂ©rapeutique. En perspective, nous tenterons d’étudier les effets du silencing d’ASPM sur la voie de signalisation la plus dĂ©rĂ©gulĂ©e (pRB / E2F ou PI3K / AKT). Enfin, nous Ă©tudierons le rĂŽle potentiel de cette protĂ©ine dans le contrĂŽle du cycle cellulaire, et, in fine la mise en Ă©vidence de ses partenaires

    Etude par arn interférence de l'expression du gÚne aspm dans les cellules souches tumorales des gliomes de haut grade

    No full text
    Les gliomes sont les tumeurs cĂ©rĂ©brales primitives les plus frĂ©quentes de l adulte. Le glioblastome (grade IV) en est la forme la plus agressive, caractĂ©risĂ© par sa rĂ©sistance aux traitements actuels (chirurgie, chimiothĂ©rapie et radiothĂ©rapie). La mortalitĂ© de cette pathologie est quasi constante (survie mĂ©diane de 15 mois), ce qui justifie l importance de dĂ©couvrir de nouvelles cibles thĂ©rapeutiques. Le challenge est d'arriver Ă  identifier des marqueurs spĂ©cifiques pour proposer un schĂ©ma thĂ©rapeutique alignant des stratĂ©gies de thĂ©rapies ciblĂ©es qui vont amĂ©liorer la prise en charge clinique, la survie globale et la survie sans progression des patients atteints de ces pathologies. Deux axes sont au centre des recherches fondamentales, translationnelles et cliniques. Le premier axe se dĂ©finit autour du dĂ©veloppement de molĂ©cules inhibitrices des voies de signalisation et le second autour du concept de cellules souches tumorales (CST) de glioblastomes (GBM) dĂ©couvertes rĂ©cemment dans le cerveau et qui rĂ©volutionnent la conception de la transformation tumorale.ASPM (Abnormal Spindle Like MicrocĂ©phaly Associated) est une cible candidate pertinente susceptible de participer au dĂ©veloppement des gliomes (Horvath et al., 2007 ; Hagmann et al., 2008). Cette protĂ©ine rĂ©gule la prolifĂ©ration des neuroblastes, elle est fortement exprimĂ©e au stade embryonnaire, mais, reste faiblement exprimĂ©e dans le cerveau adulte. Par ailleurs, ASPM est impliquĂ©e dans divers processus de cancĂ©risation (surexprimĂ©e dans les cancers du sein, du foie et du cerveau ), toute fois, le mĂ©canisme responsable de cette dĂ©rĂ©gulation n est pas encore bien caractĂ©risĂ©.Nos Ă©tudes menĂ©es sur une sĂ©rie de 169 gliomes humains, sĂ©lectionnĂ©s Ă  partir de notre cohorte de patients, montrent que le gĂšne ASPM est un marqueur de la progression vers la malignitĂ©, les grades les plus Ă©levĂ©s exprimant le plus fortement ASPM. En outre, nous avons Ă©galement montrĂ© que le niveau des transcrits d ASPM est augmentĂ© dans les rĂ©cidives de gliomes et qu en in vitro, ASPM contrĂŽle la formation des gliomasphĂšres (CST de GBM) avec une augmentation de l expression de ses transcrits dans les cultures in vitro au fil des passages. En continuitĂ© de ces observations, nous avons alors dĂ©veloppĂ© un sh-miR-RNA spĂ©cifique d ASPM permettant l extinction post-transcriptionnelle de ce gĂšne. Les rĂ©sultats obtenus in vitro montrent que la perte d expression d ASPM conduit Ă  un arrĂȘt de la prolifĂ©ration et aboutit Ă  une mort cellulaire massive.Actuellement, des modĂšles de greffe de gliomasphĂšres chez la souris (orthotopique) sont en cours de dĂ©veloppement pour confirmer les effets observĂ©s in vitro et vĂ©rifier in vivo la validitĂ© de notre approche thĂ©rapeutique. En perspective, nous tenterons d Ă©tudier les effets du silencing d ASPM sur la voie de signalisation la plus dĂ©rĂ©gulĂ©e (pRB / E2F ou PI3K / AKT). Enfin, nous Ă©tudierons le rĂŽle potentiel de cette protĂ©ine dans le contrĂŽle du cycle cellulaire, et, in fine la mise en Ă©vidence de ses partenairesGlioblastoma (GBM) is the most frequent and aggressive form of primary brain tumors in adults; it is characterized by its resistance to current treatments (surgery, chemotherapy and radiotherapy). The prognosis is grim with a median survival of only 15 months underlining the importance to develop new therapeutic strategies. The recent development of the tumor stem cell (TSC) concept in hemopathies has been secondarily applied to gliomas with the identification of subpopulations of GBM cells which express neural stem cell markers and fulfill the criteria for stemness. Some evidences also suggest that this subpopulation could play a primary role in resistance to radio- and chemotherapy.ASPM (Abnormal Spindle Like Microcephaly Associated) is a protein regulating the proliferation of neuroblasts, highly expressed in the embryonic stage but weakly expressed in the adult brain. Preliminary reports suggesting that it could be involved in the development of gliomas (Horvath et al., 2007, Hagemann et al., 2008) prompted us to analyze further the role of this protein, focusing on its potential as a relevant candidate therapeutic target. In a series of 175 gliomas samples of various grades, we found that ASPM mRNA expression was strongly correlated with increasing tumor grade. We also found that ASPM expression increased at recurrence when compared to the initial lesion. Subsequently, we could demonstrate in vitro and in vivo that ASPM expression also increased over serial passages in gliomaspheres and in a mouse glioma xenograft model. In a therapeutic perspective, the effect of lentivirus-mediated shRNA post-transcriptional silencing of ASPM was evaluated in two different gliomasphere models and a dramatic proliferation arrest and cell death was observed. Taken together, these data suggest that ASPM is involved in the malignant progression of gliomas, possibly through expansion of a cancer stem cell compartment, and could be an attractive therapeutic target in glioblastoma multiforme.Another potential candidate tumor stem cell target in glioma is the sonic hedgehog pathway (hedgehog-Gli) which is required for GBM growth and stem cell expansion. In a collaborative study, it was found that NANOG, a transcription factor critically involved with self-renewal of undifferentiated embryonic stem cells, modulates gliomasphere clonogenicity, CD133+ stem cell behavior and proliferation. NANOG was regulated by hedgehog-Gli signalling and was essential for GBM tumourigenicity in orthotopic xenografts suggesting that it could also be a useful potential therapeutic target.Conclusions: Accumulating evidences suggest that tumor stem cells play an important role in the oncogenesis of gliomas and in their resistance to treatment. Our data support this concept and suggest that specific stemness markers may become useful targets to improve treatment of this devastating disease.PARIS11-SCD-Bib. Ă©lectronique (914719901) / SudocSudocFranceF

    NANOG regulates glioma stem cells and is essential in vivo acting in a cross-functional network with GLI1 and p53

    No full text
    A cohort of genes associated with embryonic stem (ES) cell behaviour, including NANOG, are expressed in a number of human cancers. They form an ES-like signature we first described in glioblastoma multiforme (GBM), a highly invasive and incurable brain tumour. We have also shown that HEDGEHOG-GLI (HH-GLI) signalling is required for GBM growth, stem cell expansion and the expression of this (ES)-like stemness signature. Here, we address the function of NANOG in human GBMs and its relationship with HH-GLI activity. We find that NANOG modulates gliomasphere clonogenicity, CD133+ stem cell cell behavior and proliferation, and is regulated by HH-GLI signalling. However, GLI1 also requires NANOG activity forming a positive loop, which is negatively controlled by p53 and vice versa. NANOG is essential for GBM tumourigenicity in orthotopic xenografts and it is epistatic to HH-GLI activity. Our data establish NANOG as a novel HH-GLI mediator essential for GBMs. We propose that this function is conserved and that tumour growth and stem cell behaviour rely on the status of a functional GLI1-NANOG-p53 network

    Barriers to the conduct of randomised clinical trials within all disease areas

    No full text
    International audienceBackground. Randomised clinical trials are key to advancing medical knowledge and to enhancing patient care, but major barriers to their conduct exist. The present paper presents some of these barriers.Methods. We performed systematic literature searches and internal European Clinical Research Infrastructure Network (ECRIN) communications during face-to-face meetings and telephone conferences from 2013 to 2017 within the context of the ECRIN Integrating Activity (ECRIN-IA) project.Results. The following barriers to randomised clinical trials were identified: inadequate knowledge of clinical research and trial methodology; lack of funding; excessive monitoring; restrictive privacy law and lack of transparency; complex regulatory requirements; and inadequate infrastructures. There is a need for more pragmatic randomised clinical trials conducted with low risks of systematic and random errors, and multinational cooperation is essential.Conclusions. The present paper presents major barriers to randomised clinical trials. It also underlines the value of using a pan-European-distributed infrastructure to help investigators overcome barriers for multi-country trials in any disease area
    corecore