Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Alexandre Puissant is active.

Publication


Featured researches published by Alexandre Puissant.


Cancer Discovery | 2013

Targeting MYCN in Neuroblastoma by BET Bromodomain Inhibition

Alexandre Puissant; Stacey M. Frumm; Gabriela Alexe; Christopher F. Bassil; Jun Qi; Yvan Chanthery; Erin A. Nekritz; Rhamy Zeid; William Clay Gustafson; Patricia Greninger; Matthew J Garnett; Ultan McDermott; Cyril H. Benes; Andrew L. Kung; William A. Weiss; James E. Bradner; Kimberly Stegmaier

Bromodomain inhibition comprises a promising therapeutic strategy in cancer, particularly for hematologic malignancies. To date, however, genomic biomarkers to direct clinical translation have been lacking. We conducted a cell-based screen of genetically defined cancer cell lines using a prototypical inhibitor of BET bromodomains. Integration of genetic features with chemosensitivity data revealed a robust correlation between MYCN amplification and sensitivity to bromodomain inhibition. We characterized the mechanistic and translational significance of this finding in neuroblastoma, a childhood cancer with frequent amplification of MYCN. Genome-wide expression analysis showed downregulation of the MYCN transcriptional program accompanied by suppression of MYCN transcription. Functionally, bromodomain-mediated inhibition of MYCN impaired growth and induced apoptosis in neuroblastoma. BRD4 knockdown phenocopied these effects, establishing BET bromodomains as transcriptional regulators of MYCN. BET inhibition conferred a significant survival advantage in 3 in vivo neuroblastoma models, providing a compelling rationale for developing BET bromodomain inhibitors in patients with neuroblastoma.


Cancer Research | 2010

Resveratrol Promotes Autophagic Cell Death in Chronic Myelogenous Leukemia Cells via JNK-Mediated p62/SQSTM1 Expression and AMPK Activation

Alexandre Puissant; Guillaume Robert; Nina Fenouille; Frederic Luciano; Jill-Patrice Cassuto; Sophie Raynaud; Patrick Auberger

Autophagy that is induced by starvation or cellular stress can enable cancer cell survival by sustaining energy homeostasis and eliminating damaged organelles and proteins. In response to stress, cancer cells have been reported to accumulate the protein p62/SQSTM1 (p62), but its role in the regulation of autophagy is controversial. Here, we report that the plant phytoalexin resveratrol (RSV) triggers autophagy in imatinib-sensitive and imatinib-resistant chronic myelogenous leukemia (CML) cells via JNK-dependent accumulation of p62. JNK inhibition or p62 knockdown prevented RSV-mediated autophagy and antileukemic effects. RSV also stimulated AMPK, thereby inhibiting the mTOR pathway. AMPK knockdown or mTOR overexpression impaired RSV-induced autophagy but not JNK activation. Lastly, p62 expression and autophagy in CD34+ progenitors from patients with CML was induced by RSV, and disrupting autophagy protected CD34+ CML cells from RSV-mediated cell death. We concluded that RSV triggered autophagic cell death in CML cells via both JNK-mediated p62 overexpression and AMPK activation. Our findings show that the JNK and AMPK pathways can cooperate to eliminate CML cells via autophagy.


Cell Death and Disease | 2011

Metformin inhibits melanoma development through autophagy and apoptosis mechanisms

Tomic T; Botton T; Michaël Cerezo; Guillaume Robert; Frederic Luciano; Alexandre Puissant; Gounon P; Allegra M; Corine Bertolotto; Bereder Jm; Tartare-Deckert S; Bahadoran P; Patrick Auberger; Robert Ballotti; Stéphane Rocchi

Metformin is the most widely used antidiabetic drug because of its proven efficacy and limited secondary effects. Interestingly, recent studies have reported that metformin can block the growth of different tumor types. Here, we show that metformin exerts antiproliferative effects on melanoma cells, whereas normal human melanocytes are resistant to these metformin-induced effects. To better understand the basis of this antiproliferative effect of metformin in melanoma, we characterized the sequence of events underlying metformin action. We showed that 24 h metformin treatment induced a cell cycle arrest in G0/G1 phases, while after 72 h, melanoma cells underwent autophagy as demonstrated by electron microscopy, immunochemistry, and by quantification of the autolysosome-associated LC3 and Beclin1 proteins. In addition, 96 h post metformin treatment we observed robust apoptosis of melanoma cells. Interestingly, inhibition of autophagy by knocking down LC3 or ATG5 decreased the extent of apoptosis, and suppressed the antiproliferative effect of metformin on melanoma cells, suggesting that apoptosis is a consequence of autophagy. The relevance of these observations were confirmed in vivo, as we showed that metformin treatment impaired the melanoma tumor growth in mice, and induced autophagy and apoptosis markers. Taken together, our data suggest that metformin has an important impact on melanoma growth, and may therefore be beneficial in patients with melanoma.


Leukemia | 2012

The dual mTORC1 and mTORC2 inhibitor AZD8055 has anti-tumor activity in acute myeloid leukemia.

Lise Willems; Nicolas Chapuis; Alexandre Puissant; Thiago Trovati Maciel; Alexa S. Green; Nathalie Jacque; Christine Vignon; Sophie Park; S Guichard; O Herault; Aurélie Fricot; Olivier Hermine; Ivan C. Moura; Patrick Auberger; Norbert Ifrah; Francois Dreyfus; D Bonnet; Catherine Lacombe; Patrick Mayeux; Didier Bouscary; Jerome Tamburini

The serine/threonine kinase mammalian target of rapamycin (mTOR) is crucial for cell growth and proliferation, and is constitutively activated in primary acute myeloid leukemia (AML) cells, therefore representing a major target for drug development in this disease. We show here that the specific mTOR kinase inhibitor AZD8055 blocked mTORC1 and mTORC2 signaling in AML. Particularly, AZD8055 fully inhibited multisite eIF4E-binding protein 1 phosphorylation, subsequently blocking protein translation, which was in contrast to the effects of rapamycin. In addition, the mTORC1-dependent PI3K/Akt feedback activation was fully abrogated in AZD8055-treated AML cells. Significantly, AZD8055 decreased AML blast cell proliferation and cell cycle progression, reduced the clonogenic growth of leukemic progenitors and induced caspase-dependent apoptosis in leukemic cells but not in normal immature CD34+ cells. Interestingly, AZD8055 strongly induced autophagy, which may be either protective or cell death inducing, depending on concentration. Finally, AZD8055 markedly increased the survival of AML transplanted mice through a significant reduction of tumor growth, without apparent toxicity. Our current results strongly suggest that AZD8055 should be tested in AML patients in clinical trials.


Cell Death & Differentiation | 2013

Sestrin2 integrates Akt and mTOR signaling to protect cells against energetic stress-induced death

Issam Ben-Sahra; Dirat B; Kathiane Laurent; Alexandre Puissant; Patrick Auberger; Jean François Tanti; Frédéric Bost

The phosphoinositide-3 kinase/Akt (PI3K/Akt) pathway has a central role in cancer cell metabolism and proliferation. More importantly, it is one of the cardinal pro-survival pathways mediating resistance to apoptosis. The role of Akt in response to an energetic stress is presently unclear. Here, we show that Sestrin2 (Sesn2), also known as Hi95, a p53 target gene that protects cells against oxidative and genotoxic stresses, participates in the protective role of Akt in response to an energetic stress induced by 2-deoxyglucose (2-DG). Sesn2 is upregulated in response to an energetic stress such as 2-DG and metformin, and mediates the inhibition of mammalian target of rapamycin (mTOR), the major cellular regulator of energy metabolism. The increase of Sesn2 is independent of p53 but requires the anti-apoptotic pathway, PI3K/Akt. Inhibition of Akt, as well as loss of Sesn2, sensitizes cells to 2-DG-induced apoptosis. In addition, the rescue of Sesn2 partially reverses the pro-apoptotic effects of 2-DG. In conclusion, we identify Sesn2 as a new energetic stress sensor, which appears to be protective against energetic stress-induced apoptosis that integrates the pro-survival function of Akt and the negative regulation of mTOR.


PLOS ONE | 2009

Acadesine Kills Chronic Myelogenous Leukemia (CML) Cells through PKC-Dependent Induction of Autophagic Cell Death

Guillaume Robert; Issam Ben Sahra; Alexandre Puissant; Pascal Colosetti; Nathalie Belhacene; Pierre Gounon; Paul Hofman; Frédéric Bost; Jill Patrice Cassuto; Patrick Auberger

CML is an hematopoietic stem cell disease characterized by the t(9;22) (q34;q11) translocation encoding the oncoprotein p210BCR-ABL. The effect of acadesine (AICAR, 5-Aminoimidazole-4-carboxamide-1-β-D-ribofuranoside) a compound with known antileukemic effect on B cell chronic lymphoblastic leukemia (B-CLL) was investigated in different CML cell lines. Acadesine triggered loss of cell metabolism in K562, LAMA-84 and JURL-MK1 and was also effective in killing imatinib-resistant K562 cells and Ba/F3 cells carrying the T315I-BCR-ABL mutation. The anti-leukemic effect of acadesine did not involve apoptosis but required rather induction of autophagic cell death. AMPK knock-down by Sh-RNA failed to prevent the effect of acadesine, indicating an AMPK-independent mechanism. The effect of acadesine was abrogated by GF109203X and Ro-32-0432, both inhibitor of classical and new PKCs and accordingly, acadesine triggered relocation and activation of several PKC isoforms in K562 cells. In addition, this compound exhibited a potent anti-leukemic effect in clonogenic assays of CML cells in methyl cellulose and in a xenograft model of K562 cells in nude mice. In conclusion, our work identifies an original and unexpected mechanism by which acadesine triggers autophagic cell death through PKC activation. Therefore, in addition to its promising effects in B-CLL, acadesine might also be beneficial for Imatinib-resistant CML patients.


The FASEB Journal | 2008

Imatinib mesylate-resistant human chronic myelogenous leukemia cell lines exhibit high sensitivity to the phytoalexin resveratrol

Alexandre Puissant; Sébastien Grosso; Arnaud Jacquel; Nathalie Belhacene; Pascal Colosetti; Jill-Patrice Cassuto; Patrick Auberger

Imatinib is successfully used in the treat ment of chronic myelogenous leukemia (CML), and the main mechanisms of resistance in refractory patients are now partially understood. In the present study, we investigated the mechanism of action of resveratrol in imatinib‐sensitive (IM‐S) and ‐resistant (IM‐R) CML cell lines. Resveratrol induced loss of viability and apopto sis in IM‐S and IM‐R in a time‐ and dose‐dependent fashion. Inhibition of cell viability was detected for concentrations of resveratrol as low as 5 μ,M, and the IC50 values for viability, clonogenic assays, apoptosis, and erythroid differentiation were in the 10–25 μ,M range. The effect of imatinib and resveratrol was additive in IM‐S but not in IM‐R clones in which the resveratrol effect was already maximal. The effect of resveratrol on apoptosis was partially rescued by zVAD‐ fmk, suggesting a caspase‐independent contribution. Resveratrol action was independent of BCR‐ABL ex pression and phosphorylation, and in agreement was additive to BCR‐ABL silencing. Finally, phytoalexin inhibited the growth of BaF3 cells expressing mutant BCR‐ABL proteins found in resistant patients, including the multiresistant T315I mutation. Our findings show that resveratrol induces apoptosis, caspase‐inde‐ pendent death, and differentiation that collectively contribute to the specific elimination of CML cells. Resveratrol should provide therapeutic benefits in IM‐R patients and in other hematopoietic malignancies.— Puissant, A., Grosso, S., Jacquel, A., Belhacene, N., Colosetti, P., Cassuto, J.‐P., Auberger, P. Imatinib mesylate‐resistant human chronic myelogenous leuke mia cell lines exhibit high sensitivity to the phytoalexin resveratrol. FASEB J. 22, 1894–1904 (2008)


Autophagy | 2009

Autophagy is an important event for megakaryocytic differentiation of the chronic myelogenous leukemia K562 cell line

Pascal Colosetti; Alexandre Puissant; Guillaume Robert; Frederic Luciano; Arnaud Jacquel; Pierre Gounon; Jill-Patrice Cassuto; Patrick Auberger

Autophagy is a highly conserved catabolic process for the elimination and recycling of organelles and macromolecules, characterized by the formation of double membrane vesicles called autophagosomes. To date, the function of autophagy in cell differentiation is poorly documented. Here, we investigated the possibility that megakaryocytic differentiation of the Chronic Myelogenous Leukemia (CML) cell line K562, a process known to be accompanied by accumulation of vacuoles inside the cells, might involve autophagy. We show using various complementary approaches that the combination of the phorbol ester PMA and the p38MAPK inhibitor SB202190 (SB) which engaged a majority of K562 cells towards the megakaryocytic lineage also triggered vacuolization and autophagy. The combination of PMA+SB appears to induces both increase in autophagic fluxes and an autophagic degradation blockage. Induction of autophagy was accompanied also by increased expression of Beclin-1 and p62/SQSTM1 and was found to precede the onset of megakaryocytic differentiation. Moreover, knock-down of LC3 and Beclin-1 by specific siRNAs impaired PMA+SB-mediated vacuolization, LC3-II accumulation and megakaryocytic differentiation, as well. To the best of our knowledge, this is the first description that induction of autophagy is involved in megakaryocytic differentiation of K562 CML cells.


Cancer Cell | 2014

SYK Is a Critical Regulator of FLT3 in Acute Myeloid Leukemia

Alexandre Puissant; Nina Fenouille; Gabriela Alexe; Yana Pikman; Christopher F. Bassil; Swapnil Mehta; Jinyan Du; Julhash U. Kazi; Frederic Luciano; Lars Rönnstrand; Andrew L. Kung; Ilene Galinsky; Richard Stone; Daniel J. DeAngelo; Michael T. Hemann; Kimberly Stegmaier

Cooperative dependencies between mutant oncoproteins and wild-type proteins are critical in cancer pathogenesis and therapy resistance. Although spleen tyrosine kinase (SYK) has been implicated in hematologic malignancies, it is rarely mutated. We used kinase activity profiling to identify collaborators of SYK in acute myeloid leukemia (AML) and determined that FMS-like tyrosine kinase 3 (FLT3) is transactivated by SYK via direct binding. Highly activated SYK is predominantly found in FLT3-ITD positive AML and cooperates with FLT3-ITD to activate MYC transcriptional programs. FLT3-ITD AML cells are more vulnerable to SYK suppression than FLT3 wild-type counterparts. In a FLT3-ITD in vivo model, SYK is indispensable for myeloproliferative disease (MPD) development, and SYK overexpression promotes overt transformation to AML and resistance to FLT3-ITD-targeted therapy.


Molecular Cancer Therapeutics | 2009

Gene expression profiling of imatinib and PD166326-resistant CML cell lines identifies Fyn as a gene associated with resistance to BCR-ABL inhibitors

Sébastien Grosso; Alexandre Puissant; Maeva Dufies; Pascal Colosetti; Arnaud Jacquel; Kevin Lebrigand; Pascal Barbry; Marcel Deckert; Jill Patrice Cassuto; Bernard Mari; Patrick Auberger

Imatinib is used to treat chronic myelogenous leukemia (CML), but resistance develops in all phases of this disease. The purpose of the present study was to identify the mode of resistance of newly derived imatinib-resistant (IM-R) and PD166326-resistant (PD-R) CML cells. IM-R and PD-R clones exhibited an increase in viability and a decrease in caspase activation in response to various doses of imatinib and PD166326, respectively, as compared with parental K562 cells. Resistance involved neither mutations in BCR-ABL nor increased BCR-ABL, MDR1 or Lyn expression, all known modes of resistance. To gain insight into the resistance mechanisms, we used pangenomic microarrays and identified 281 genes modulated in parental versus IM-R and PD-R cells. The gene signature was similar for IM-R and PD-R cells, accordingly with the cross-sensitivity observed for both inhibitors. These genes were functionally associated with pathways linked to development, cell adhesion, cell growth, and the JAK-STAT cascade. Especially relevant were the increased expression of the tyrosine kinases AXL and Fyn as well as CD44 and HMGA2. Small interfering RNA experiments and pharmacologic approaches identified FYN as a candidate for resistance to imatinib. Our findings provide a comprehensive picture of the transcriptional events associated with imatinib and PD166326 resistance and identify Fyn as a new potential target for therapeutic intervention in CML. [Mol Cancer Ther 2009;8(7):1924–33]

Collaboration


Dive into the Alexandre Puissant's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nina Fenouille

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Pascal Colosetti

University of Nice Sophia Antipolis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maeva Dufies

University of Nice Sophia Antipolis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sophie Raynaud

University of Nice Sophia Antipolis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael T. Hemann

Massachusetts Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge