Network


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

Hotspot


Dive into the research topics where Xavier Bisteau is active.

Publication


Featured researches published by Xavier Bisteau.


Cancers | 2014

The Complex Relationship between Liver Cancer and the Cell Cycle: A Story of Multiple Regulations

Xavier Bisteau; Matias J. Caldez; Philipp Kaldis

The liver acts as a hub for metabolic reactions to keep a homeostatic balance during development and growth. The process of liver cancer development, although poorly understood, is related to different etiologic factors like toxins, alcohol, or viral infection. At the molecular level, liver cancer is characterized by a disruption of cell cycle regulation through many molecular mechanisms. In this review, we focus on the mechanisms underlying the lack of regulation of the cell cycle during liver cancer, focusing mainly on hepatocellular carcinoma (HCC). We also provide a brief summary of novel therapies connected to cell cycle regulation.


Proceedings of the National Academy of Sciences of the United States of America | 2015

A haploid genetic screen identifies the G1/S regulatory machinery as a determinant of Wee1 inhibitor sensitivity

Anne Margriet Heijink; Vincent A. Blomen; Xavier Bisteau; Fabian Degener; Felipe Yu Matsushita; Philipp Kaldis; Floris Foijer; Marcel A. T. M. van Vugt

Significance Inhibition of Wee1 is considered an attractive anticancer therapy for TP53 mutant tumors. However, additional factors besides p53 inactivation may determine Wee1 inhibitor sensitivity, which we searched for using unbiased functional genetic screening. We discovered that the mutational status of several S-phase genes, including CDK2, determines the cytotoxicity induced by Wee1 inhibition. Notably, we found that Wee1 inhibition induces two distinct phenotypes: accumulation of DNA damage in S phase and karyokinesis/cytokinesis failure during mitosis. Stable depletion of S-phase genes only reversed the formation of DNA damage, but did not rescue karyokinesis/cytokinesis failure upon Wee1 inhibition. Thus, inactivation of nonessential S-phase genes can overcome Wee1 inhibitor resistance, while allowing the survival of genomically instable cancer cells. The Wee1 cell cycle checkpoint kinase prevents premature mitotic entry by inhibiting cyclin-dependent kinases. Chemical inhibitors of Wee1 are currently being tested clinically as targeted anticancer drugs. Wee1 inhibition is thought to be preferentially cytotoxic in p53-defective cancer cells. However, TP53 mutant cancers do not respond consistently to Wee1 inhibitor treatment, indicating the existence of genetic determinants of Wee1 inhibitor sensitivity other than TP53 status. To optimally facilitate patient selection for Wee1 inhibition and uncover potential resistance mechanisms, identification of these currently unknown genes is necessary. The aim of this study was therefore to identify gene mutations that determine Wee1 inhibitor sensitivity. We performed a genome-wide unbiased functional genetic screen in TP53 mutant near-haploid KBM-7 cells using gene-trap insertional mutagenesis. Insertion site mapping of cells that survived long-term Wee1 inhibition revealed enrichment of G1/S regulatory genes, including SKP2, CUL1, and CDK2. Stable depletion of SKP2, CUL1, or CDK2 or chemical Cdk2 inhibition rescued the γ-H2AX induction and abrogation of G2 phase as induced by Wee1 inhibition in breast and ovarian cancer cell lines. Remarkably, live cell imaging showed that depletion of SKP2, CUL1, or CDK2 did not rescue the Wee1 inhibition-induced karyokinesis and cytokinesis defects. These data indicate that the activity of the DNA replication machinery, beyond TP53 mutation status, determines Wee1 inhibitor sensitivity, and could serve as a selection criterion for Wee1-inhibitor eligible patients. Conversely, loss of the identified S-phase genes could serve as a mechanism of acquired resistance, which goes along with development of severe genomic instability.


Haematologica | 2015

Hematopoiesis specific loss of Cdk2 and Cdk4 results in increased erythrocyte size and delayed platelet recovery following stress

Senthil Raja Jayapal; Chelsia Qiuxia Wang; Xavier Bisteau; Matias J. Caldez; Shuhui Lim; Vinay Tergaonkar; Motomi Osato; Philipp Kaldis

Mouse knockouts of Cdk2 and Cdk4 are individually viable whereas the double knockouts are embryonic lethal due to heart defects, and this precludes the investigation of their overlapping roles in definitive hematopoiesis. Here we use a conditional knockout mouse model to investigate the effect of combined loss of Cdk2 and Cdk4 in hematopoietic cells. Cdk2fl/flCdk4−/−vavCre mice are viable but displayed a significant increase in erythrocyte size. Cdk2fl/flCdk4−/−vavCre mouse bone marrow exhibited reduced phosphorylation of the retinoblastoma protein and reduced expression of E2F target genes such as cyclin A2 and Cdk1. Erythroblasts lacking Cdk2 and Cdk4 displayed a lengthened G1 phase due to impaired phosphorylation of the retinoblastoma protein. Deletion of the retinoblastoma protein rescued the increased size displayed by erythrocytes lacking Cdk2 and Cdk4, indicating that the retinoblastoma/Cdk2/Cdk4 pathway regulates erythrocyte size. The recovery of platelet counts following a 5-fluorouracil challenge was delayed in Cdk2fl/flCdk4−/−vavCre mice revealing a critical role for Cdk2 and Cdk4 in stress hematopoiesis. Our data indicate that Cdk2 and Cdk4 play important overlapping roles in homeostatic and stress hematopoiesis, which need to be considered when using broad-spectrum cyclin-dependent kinase inhibitors for cancer therapy.


Science | 2018

Thermal proximity coaggregation for system-wide profiling of protein complex dynamics in cells

Chris Soon Heng Tan; Ka Diam Go; Xavier Bisteau; Lingyun Dai; Chern Han Yong; Nayana Prabhu; Mert Burak Ozturk; Yan Ting Lim; Lekshmy Sreekumar; Johan Lengqvist; Vinay Tergaonkar; Philipp Kaldis; Radoslaw M. Sobota; Pär Nordlund

Taking the heat together Many of the processes in living cells are mediated by protein complexes that dynamically assemble and dissociate depending on cellular needs. Tan et al. developed a method called thermal proximity coaggregation (TPCA) to monitor the dynamics of native protein complexes inside cells (see the Perspective by Li et al.). The method is based on the idea that proteins within a complex will coaggregate upon heat denaturation. It uses a previously described cellular shift assay to determine melting curves for thousands of proteins and assigns a TPCA signature on the basis of similarity between the curves. The method was validated by detection of many known protein complexes. It identified cell-specific interactions in six cell lines, highlighting the potential for identifying protein complexes that are modulated by disease. Science, this issue p. 1170; see also p. 1105 A readily deployable approach for system-wide intracellular studies of protein complex dynamics in nonengineered cells and tissues is discussed. Proteins differentially interact with each other across cellular states and conditions, but an efficient proteome-wide strategy to monitor them is lacking. We report the application of thermal proximity coaggregation (TPCA) for high-throughput intracellular monitoring of protein complex dynamics. Significant TPCA signatures observed among well-validated protein-protein interactions correlate positively with interaction stoichiometry and are statistically observable in more than 350 annotated human protein complexes. Using TPCA, we identified many complexes without detectable differential protein expression, including chromatin-associated complexes, modulated in S phase of the cell cycle. Comparison of six cell lines by TPCA revealed cell-specific interactions even in fundamental cellular processes. TPCA constitutes an approach for system-wide studies of protein complexes in nonengineered cells and tissues and might be used to identify protein complexes that are modulated in diseases.


PLOS Genetics | 2016

Loss of the Greatwall Kinase Weakens the Spindle Assembly Checkpoint.

M. Kasim Diril; Xavier Bisteau; Mayumi Kitagawa; Matias J. Caldez; Sheena Wee; Jayantha Gunaratne; Sang Hyun Lee; Philipp Kaldis

The Greatwall kinase/Mastl is an essential gene that indirectly inhibits the phosphatase activity toward mitotic Cdk1 substrates. Here we show that although Mastl knockout (MastlNULL) MEFs enter mitosis, they progress through mitosis without completing cytokinesis despite the presence of misaligned chromosomes, which causes chromosome segregation defects. Furthermore, we uncover the requirement of Mastl for robust spindle assembly checkpoint (SAC) maintenance since the duration of mitotic arrest caused by microtubule poisons in MastlNULL MEFs is shortened, which correlates with premature disappearance of the essential SAC protein Mad1 at the kinetochores. Notably, MastlNULL MEFs display reduced phosphorylation of a number of proteins in mitosis, which include the essential SAC kinase MPS1. We further demonstrate that Mastl is required for multi-site phosphorylation of MPS1 as well as robust MPS1 kinase activity in mitosis. In contrast, treatment of MastlNULL cells with the phosphatase inhibitor okadaic acid (OKA) rescues the defects in MPS1 kinase activity, mislocalization of phospho-MPS1 as well as Mad1 at the kinetochore, and premature SAC silencing. Moreover, using in vitro dephosphorylation assays, we demonstrate that Mastl promotes persistent MPS1 phosphorylation by inhibiting PP2A/B55-mediated MPS1 dephosphorylation rather than affecting Cdk1 kinase activity. Our findings establish a key regulatory function of the Greatwall kinase/Mastl->PP2A/B55 pathway in preventing premature SAC silencing.


American Journal of Human Genetics | 2017

CDK10 Mutations in Humans and Mice Cause Severe Growth Retardation, Spine Malformations, and Developmental Delays

Christian Windpassinger; Juliette Piard; Carine Bonnard; Majid Alfadhel; Shuhui Lim; Xavier Bisteau; Stéphane Blouin; Nur’Ain B. Ali; Alvin Yu Jin Ng; Hao Lu; Sumanty Tohari; S. Zakiah A. Talib; Noémi van Hul; Matias J. Caldez; Lionel Van Maldergem; Gökhan Yigit; Hülya Kayserili; Sameh A. Youssef; Vincenzo Coppola; Alain de Bruin; Lino Tessarollo; Hyungwon Choi; Verena Rupp; Katharina M. Roetzer; Paul Roschger; Klaus Klaushofer; Janine Altmüller; Sudipto Roy; Byrappa Venkatesh; Rudolf Ganger

In five separate families, we identified nine individuals affected by a previously unidentified syndrome characterized by growth retardation, spine malformation, facial dysmorphisms, and developmental delays. Using homozygosity mapping, array CGH, and exome sequencing, we uncovered bi-allelic loss-of-function CDK10 mutations segregating with this disease. CDK10 is a protein kinase that partners with cyclin M to phosphorylate substrates such as ETS2 and PKN2 in order to modulate cellular growth. To validate and model the pathogenicity of these CDK10 germline mutations, we generated conditional-knockout mice. Homozygous Cdk10-knockout mice died postnatally with severe growth retardation, skeletal defects, and kidney and lung abnormalities, symptoms that partly resemble the diseases effect in humans. Fibroblasts derived from affected individuals and Cdk10-knockout mouse embryonic fibroblasts (MEFs) proliferated normally; however, Cdk10-knockout MEFs developed longer cilia. Comparative transcriptomic analysis of mutant and wild-type mouse organs revealed lipid metabolic changes consistent with growth impairment and altered ciliogenesis in the absence of CDK10. Our results document the CDK10 loss-of-function phenotype and point to a function for CDK10 in transducing signals received at the primary cilia to sustain embryonic and postnatal development.


Cell Cycle | 2016

Cyclin A2 regulates erythrocyte morphology and numbers

Senthil Raja Jayapal; Heather Yin-Kuan Ang; Chelsia Qiuxia Wang; Xavier Bisteau; Matias J. Caldez; Gan Xiao Xuan; Weimiao Yu; Vinay Tergaonkar; Motomi Osato; Bing Lim; Philipp Kaldis

ABSTRACT Cyclin A2 is an essential gene for development and in haematopoietic stem cells and therefore its functions in definitive erythropoiesis have not been investigated. We have ablated cyclin A2 in committed erythroid progenitors in vivo using erythropoietin receptor promoter-driven Cre, which revealed its critical role in regulating erythrocyte morphology and numbers. Erythroid-specific cyclin A2 knockout mice are viable but displayed increased mean erythrocyte volume and reduced erythrocyte counts, as well as increased frequency of erythrocytes containing Howell-Jolly bodies. Erythroblasts lacking cyclin A2 displayed defective enucleation, resulting in reduced production of enucleated erythrocytes and increased frequencies of erythrocytes containing nuclear remnants. Deletion of the Cdk inhibitor p27Kip1 but not Cdk2, ameliorated the erythroid defects resulting from deficiency of cyclin A2, confirming the critical role of cyclin A2/Cdk activity in erythroid development. Loss of cyclin A2 in bone marrow cells in semisolid culture prevented the formation of BFU-E but not CFU-E colonies, uncovering its essential role in BFU-E function. Our data unveils the critical functions of cyclin A2 in regulating mammalian erythropoiesis.


Oncogene | 2018

Premature activation of Cdk1 leads to mitotic events in S phase and embryonic lethality

Radoslaw Szmyd; Joanna Niska-Blakie; M. Kasim Diril; Patrícia Renck Nunes; Konstantinos Tzelepis; Aurélie Lacroix; Noémi van Hul; Lih-Wen Deng; Joao Matos; Oliver Dreesen; Xavier Bisteau; Philipp Kaldis

Cell cycle regulation, especially faithful DNA replication and mitosis, are crucial to maintain genome stability. Cyclin-dependent kinase (CDK)/cyclin complexes drive most processes in cellular proliferation. In response to DNA damage, cell cycle surveillance mechanisms enable normal cells to arrest and undergo repair processes. Perturbations in genomic stability can lead to tumor development and suggest that cell cycle regulators could be effective targets in anticancer therapy. However, many clinical trials ended in failure due to off-target effects of the inhibitors used. Here, we investigate in vivo the importance of WEE1- and MYT1-dependent inhibitory phosphorylation of mammalian CDK1. We generated Cdk1AF knockin mice, in which two inhibitory phosphorylation sites are replaced by the non-phosphorylatable amino acids T14A/Y15F. We uncovered that monoallelic expression of CDK1AF is early embryonic lethal in mice and induces S phase arrest accompanied by γH2AX and DNA damage checkpoint activation in mouse embryonic fibroblasts (MEFs). The chromosomal fragmentation in Cdk1AF MEFs does not rely on CDK2 and is partly caused by premature activation of MUS81-SLX4 structure-specific endonuclease complexes, as well as untimely onset of chromosome condensation followed by nuclear lamina disassembly. We provide evidence that tumor development in liver expressing CDK1AF is inhibited. Interestingly, the regulatory mechanisms that impede cell proliferation in CDK1AF expressing cells differ partially from the actions of the WEE1 inhibitor, MK-1775, with p53 expression determining the sensitivity of cells to the drug response. Thus, our work highlights the importance of improved therapeutic strategies for patients with various cancer types and may explain why some patients respond better to WEE1 inhibitors.


Cell | 2018

Modulation of Protein-Interaction States through the Cell Cycle

Lingyun Dai; Tianyun Zhao; Xavier Bisteau; Wendi Sun; Nayana Prabhu; Yan Ting Lim; Radoslaw M. Sobota; Philipp Kaldis; Pär Nordlund

Global profiling of protein expression through the cell cycle has revealed subsets of periodically expressed proteins. However, expression levels alone only give a partial view of the biochemical processes determining cellular events. Using a proteome-wide implementation of the cellular thermal shift assay (CETSA) to study specific cell-cycle phases, we uncover changes of interaction states for more than 750 proteins during the cell cycle. Notably, many protein complexes are modulated in specific cell-cycle phases, reflecting their roles in processes such as DNA replication, chromatin remodeling, transcription, translation, and disintegration of the nuclear envelope. Surprisingly, only small differences in the interaction states were seen between the G1 and the G2 phase, suggesting similar hardwiring of biochemical processes in these two phases. The present work reveals novel molecular details of the cell cycle and establishes proteome-wide CETSA as a new strategy to study modulation of protein-interaction states in intact cells.


Biochemical Journal | 2016

Cdk2 catalytic activity is essential for meiotic cell division in vivo

Sangeeta Chauhan; M. Kasim Diril; Joanna H.S. Lee; Xavier Bisteau; Vanessa Manoharan; Deepak Adhikari; Chandrahas Koumar Ratnacaram; Baptiste Janela; Juliane Noffke; Florent Ginhoux; Vincenzo Coppola; Kui Liu; Lino Tessarollo; Philipp Kaldis

Collaboration


Dive into the Xavier Bisteau's collaboration.

Top Co-Authors

Avatar

Philipp Kaldis

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lingyun Dai

Nanyang Technological University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nayana Prabhu

Nanyang Technological University

View shared research outputs
Top Co-Authors

Avatar

Pär Nordlund

Nanyang Technological University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yan Ting Lim

Nanyang Technological University

View shared research outputs
Top Co-Authors

Avatar

Chelsia Qiuxia Wang

National University of Singapore

View shared research outputs
Researchain Logo
Decentralizing Knowledge