Network


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

Hotspot


Dive into the research topics where Martine F. Roussel is active.

Publication


Featured researches published by Martine F. Roussel.


Cell | 1997

Tumor Suppression at the Mouse INK4a Locus Mediated by the Alternative Reading Frame Product p19 ARF

Takehiko Kamijo; Frederique Zindy; Martine F. Roussel; Dawn E. Quelle; James R. Downing; Richard A. Ashmun; Gerard Grosveld; Charles J. Sherr

The INK4a tumor suppressor locus encodes p16INK4a, an inhibitor of cyclin D-dependent kinases, and p19ARF, an alternative reading frame protein that also blocks cell proliferation. Surprisingly, mice lacking p19ARF but expressing functional p16INK4a develop tumors early in life. Their embryo fibroblasts (MEFs) do not senesce and are transformed by oncogenic Ha-ras alone. Conversion of ARF+/+ or ARF+/- MEF strains to continuously proliferating cell lines involves loss of either p19ARF or p53. p53-mediated checkpoint control is unperturbed in ARF-null fibroblast strains, whereas p53-negative cell lines are resistant to p19ARF-induced growth arrest. Therefore, INK4a encodes growth inhibitory proteins that act upstream of the retinoblastoma protein and p53. Mutations and deletions targeting this locus in cancer cells are unlikely to be functionally equivalent.


Cell | 1985

The c-fms proto-oncogene product is related to the receptor for the mononuclear phagocyte growth factor, CSF 1

Charles J. Sherr; Carl W. Rettenmier; Rosalba Sacca; Martine F. Roussel; A. Thomas Look; E. Richard Stanley

The feline c-fms proto-oncogene product is a 170 kd glycoprotein with associated tyrosine kinase activity. This glycoprotein was expressed on mature cat macrophages from peritoneal inflammatory exudates and spleen. Similarly, the receptor for the murine colony-stimulating factor, CSF-1, is restricted to cells of the mononuclear phagocytic lineage and is a 165 kd glycoprotein with an associated tyrosine kinase. Rabbit antisera to a recombinant v-fms-coded polypeptide precipitated the feline c-fms product and specifically cross-reacted with a 165 kd glycoprotein from mouse macrophages. This putative product of the murine c-fms gene exhibited an associated tyrosine kinase activity in immune complexes, specifically bound murine CSF-1, and, in the presence of the growth factor, was phosphorylated on tyrosine in membrane preparations. The murine c-fms proto-oncogene product and the CSF-1 receptor are therefore related, and possibly identical, molecules.


The EMBO Journal | 1999

The p21 Cip1 and p27 Kip1 CDK ‘inhibitors’ are essential activators of cyclin D‐dependent kinases in murine fibroblasts

Mangeng Cheng; Paul Olivier; J.Alan Diehl; Matthew L. Fero; Martine F. Roussel; James M. Roberts; Charles J. Sherr

The widely prevailing view that the cyclin‐dependent kinase inhibitors (CKIs) are solely negative regulators of cyclin‐dependent kinases (CDKs) is challenged here by observations that normal up‐regulation of cyclin D–CDK4 in mitogen‐stimulated fibroblasts depends redundantly upon p21Cip1 and p27Kip1. Primary mouse embryonic fibroblasts that lack genes encoding both p21 and p27 fail to assemble detectable amounts of cyclin D–CDK complexes, express cyclin D proteins at much reduced levels, and are unable to efficiently direct cyclin D proteins to the cell nucleus. Restoration of CKI function reverses all three defects and thereby restores cyclin D activity to normal physiological levels. In the absence of both CKIs, the severe reduction in cyclin D‐dependent kinase activity was well tolerated and had no overt effects on the cell cycle.


Cell | 1991

Colony-stimulating factor 1 regulates novel cyclins during the G1 phase of the cell cycle

Hitoshl Matsushime; Martine F. Roussel; Richard A. Ashmun; Charles J. Sherr

Three mouse cyclin-like (CYL) genes were isolated, two of which are regulated by colony-stimulating factor 1 (CSF-1) during the G1 phase of the macrophage cell cycle. CSF-1 deprivation during G1 leads to rapid degradation of CYL proteins (p36CYL) and correlates with failure to initiate DNA synthesis. However, after entering S phase, macrophages no longer require CSF-1 and can complete cell division without expressing CYL genes. During G1, p36CYL is phosphorylated and associates with a polypeptide antigenically related to p34cdc2. The timing of p36CYL expression, its rapid turnover in the absence of CSF-1, and its phosphorylation and transient binding to a cdc2-related polypeptide suggest that CYL genes may function during S phase commitment.


Nature Cell Biology | 1999

Nucleolar Arf sequesters Mdm2 and activates p53

Jason D. Weber; Laura J. Taylor; Martine F. Roussel; Charles J. Sherr; Dafna Bar-Sagi

The Ink4/Arf locus encodes two tumour-suppressor proteins, p16Ink4a and p19Arf, that govern the antiproliferative functions of the retinoblastoma and p53 proteins, respectively. Here we show that Arf binds to the product of the Mdm2 gene and sequesters it into the nucleolus, thereby preventing negative-feedback regulation of p53 by Mdm2 and leading to the activation of p53 in the nucleoplasm. Arf and Mdm2 co-localize in the nucleolus in response to activation of the oncoprotein Myc and as mouse fibroblasts undergo replicative senescence. These topological interactions of Arf and Mdm2 point towards a new mechanism for p53 activation.


Cell | 1992

Identification and properties of an atypical catalytic subunit (p34PSK-J3/cdk4) for mammalian D type G1 cyclins

Hitoshi Matsushime; Mark E. Ewen; David K. Strom; Jun-ya Kato; Steven K. Hanks; Martine F. Roussel; Charles J. Sherr

Murine D type cyclins associate with a catalytic subunit (p34PSK-J3) with properties distinct from known cyclin-dependent kinases (cdks). Mouse p34PSK-J3 shows less than 50% amino acid identity to p34cdc2, p33cdk2, and p36cdk3, lacks a PSTAIRE motif, and does not bind to p13suc1. Cyclin D1-p34PSK-J3 complexes accumulate in macrophages during G1 and decline in S phase, whereas complexes involving cyclins D2 and D3 form in proliferating T cells. Although histone H1 kinase activity is not detected in cyclin D or PSK-J3 immunoprecipitates, cyclin D-p34PSK-J3 complexes assembled in vitro stably bind and phosphorylate the retinoblastoma gene product (pRb) and an Rb-like protein (p107) but do not interact with pRb mutants that are functionally inactive. Thus, p34PSK-J3 is a cyclin D-regulated catalytic subunit that acts as an Rb (but not H1) kinase.


Molecular and Cellular Biology | 1995

Novel INK4 proteins, p19 and p18, are specific inhibitors of the cyclin D-dependent kinases CDK4 and CDK6.

Hiroshi Hirai; Martine F. Roussel; Jun-ya Kato; Richard A. Ashmun; Charles J. Sherr

Cyclin D-dependent kinases act as mitogen-responsive, rate-limiting controllers of G1 phase progression in mammalian cells. Two novel members of the mouse INK4 gene family, p19 and p18, that specifically inhibit the kinase activities of CDK4 and CDK6, but do not affect those of cyclin E-CDK2, cyclin A-CDK2, or cyclin B-CDC2, were isolated. Like the previously described human INK4 polypeptides, p16INK4a/MTS1 and p15INK4b/MTS2, mouse p19 and p18 are primarily composed of tandemly repeated ankyrin motifs, each ca. 32 amino acids in length, p19 and p18 bind directly to CDK4 and CDK6, whether untethered or in complexes with D cyclins, and can inhibit the activity of cyclin D-bound cyclin-dependent kinases (CDKs). Although neither protein interacts with D cyclins or displaces them from preassembled cyclin D-CDK complexes in vitro, both form complexes with CDKs at the expense of cyclins in vivo, suggesting that they may also interfere with cyclin-CDK assembly. In proliferating macrophages, p19 mRNA and protein are periodically expressed with a nadir in G1 phase and maximal synthesis during S phase, consistent with the possibility that INK4 proteins limit the activities of CDKs once cells exit G1 phase. However, introduction of a vector encoding p19 into mouse NIH 3T3 cells leads to constitutive p19 synthesis, inhibits cyclin D1-CDK4 activity in vivo, and induces G1 phase arrest.


Nature | 2010

Subtypes of medulloblastoma have distinct developmental origins.

Paul Gibson; Yiai Tong; Giles W. Robinson; Margaret C. Thompson; D. Spencer Currle; Christopher Eden; Tanya A. Kranenburg; Twala L. Hogg; Helen Poppleton; Julie Martin; David Finkelstein; Stanley Pounds; Aaron Weiss; Zoltan Patay; Matthew A. Scoggins; Robert J. Ogg; Yanxin Pei; Zeng-Jie Yang; Sonja N. Brun; Youngsoo Lee; Frederique Zindy; Janet C. Lindsey; Makoto M. Taketo; Frederick A. Boop; Robert A. Sanford; Amar Gajjar; Steven C. Clifford; Martine F. Roussel; Peter J. McKinnon; David H. Gutmann

Medulloblastoma encompasses a collection of clinically and molecularly diverse tumour subtypes that together comprise the most common malignant childhood brain tumour. These tumours are thought to arise within the cerebellum, with approximately 25% originating from granule neuron precursor cells (GNPCs) after aberrant activation of the Sonic Hedgehog pathway (hereafter, SHH subtype). The pathological processes that drive heterogeneity among the other medulloblastoma subtypes are not known, hindering the development of much needed new therapies. Here we provide evidence that a discrete subtype of medulloblastoma that contains activating mutations in the WNT pathway effector CTNNB1 (hereafter, WNT subtype) arises outside the cerebellum from cells of the dorsal brainstem. We found that genes marking human WNT-subtype medulloblastomas are more frequently expressed in the lower rhombic lip (LRL) and embryonic dorsal brainstem than in the upper rhombic lip (URL) and developing cerebellum. Magnetic resonance imaging (MRI) and intra-operative reports showed that human WNT-subtype tumours infiltrate the dorsal brainstem, whereas SHH-subtype tumours are located within the cerebellar hemispheres. Activating mutations in Ctnnb1 had little impact on progenitor cell populations in the cerebellum, but caused the abnormal accumulation of cells on the embryonic dorsal brainstem which included aberrantly proliferating Zic1+ precursor cells. These lesions persisted in all mutant adult mice; moreover, in 15% of cases in which Tp53 was concurrently deleted, they progressed to form medulloblastomas that recapitulated the anatomy and gene expression profiles of human WNT-subtype medulloblastoma. We provide the first evidence, to our knowledge, that subtypes of medulloblastoma have distinct cellular origins. Our data provide an explanation for the marked molecular and clinical differences between SHH- and WNT-subtype medulloblastomas and have profound implications for future research and treatment of this important childhood cancer.


Nature | 2012

Novel mutations target distinct subgroups of medulloblastoma

Giles W. Robinson; Matthew Parker; Tanya A. Kranenburg; Charles Lu; Xiang Chen; Li Ding; Timothy N. Phoenix; Erin Hedlund; Lei Wei; Xiaoyan Zhu; Nader Chalhoub; Suzanne J. Baker; Robert Huether; Richard W. Kriwacki; Natasha Curley; Radhika Thiruvenkatam; Jianmin Wang; Gang Wu; Michael Rusch; Xin Hong; Jared Becksfort; Pankaj Gupta; Jing Ma; John Easton; Bhavin Vadodaria; Arzu Onar-Thomas; Tong Lin; Shaoyi Li; Stanley Pounds; Steven W. Paugh

Medulloblastoma is a malignant childhood brain tumour comprising four discrete subgroups. Here, to identify mutations that drive medulloblastoma, we sequenced the entire genomes of 37 tumours and matched normal blood. One-hundred and thirty-six genes harbouring somatic mutations in this discovery set were sequenced in an additional 56 medulloblastomas. Recurrent mutations were detected in 41 genes not yet implicated in medulloblastoma; several target distinct components of the epigenetic machinery in different disease subgroups, such as regulators of H3K27 and H3K4 trimethylation in subgroups 3 and 4 (for example, KDM6A and ZMYM3), and CTNNB1-associated chromatin re-modellers in WNT-subgroup tumours (for example, SMARCA4 and CREBBP). Modelling of mutations in mouse lower rhombic lip progenitors that generate WNT-subgroup tumours identified genes that maintain this cell lineage (DDX3X), as well as mutated genes that initiate (CDH1) or cooperate (PIK3CA) in tumorigenesis. These data provide important new insights into the pathogenesis of medulloblastoma subgroups and highlight targets for therapeutic development.


Molecular and Cellular Biology | 2000

Cooperative Signals Governing ARF-Mdm2 Interaction and Nucleolar Localization of the Complex

Jason D. Weber; Mei-Ling Kuo; Brian Bothner; Enrico L. DiGiammarino; Richard W. Kriwacki; Martine F. Roussel; Charles J. Sherr

ABSTRACT The ARF tumor suppressor protein stabilizes p53 by antagonizing its negative regulator, Mdm2 (Hdm2 in humans). Both mouse p19 ARF and human p14 ARF bind to the central region of Mdm2 (residues 210 to 304), a segment that does not overlap with its N-terminal p53-binding domain, nuclear import or export signals, or C-terminal RING domain required for Mdm2 E3 ubiquitin ligase activity. The N-terminal 37 amino acids of mouse p19 ARF are necessary and sufficient for binding to Mdm2, localization of Mdm2 to nucleoli, and p53-dependent cell cycle arrest. Although a nucleolar localization signal (NrLS) maps within a different segment (residues 82 to 101) of the human p14 ARF protein, binding to Mdm2 and nucleolar import of ARF-Mdm2 complexes are both required for cell cycle arrest induced by either the mouse or human ARF proteins. Because many codons of mouse ARF mRNA are not recognized by the most abundant bacterial tRNAs, we synthesized ARF minigenes containing preferred bacterial codons. Using bacterially produced ARF polypeptides and chemically synthesized peptides conjugated to Sepharose, residues 1 to 14 and 26 to 37 of mouse p19 ARF were found to interact independently and cooperatively with Mdm2, while residues 15 to 25 were dispensable for binding. Paradoxically, residues 26 to 37 of mouse p19 ARF are also essential for ARF nucleolar localization in the absence of Mdm2. However, the mobilization of the p19 ARF -Mdm2 complex into nucleoli also requires a cryptic NrLS within the Mdm2 C-terminal RING domain. The Mdm2 NrLS is unmasked upon ARF binding, and its deletion prevents import of the ARF-Mdm2 complex into nucleoli. Collectively, the results suggest that ARF binding to Mdm2 induces a conformational change that facilitates nucleolar import of the ARF-Mdm2 complex and p53-dependent cell cycle arrest. Hence, the ARF-Mdm2 interaction can be viewed as bidirectional, with each protein being capable of regulating the subnuclear localization of the other.

Collaboration


Dive into the Martine F. Roussel's collaboration.

Top Co-Authors

Avatar

Charles J. Sherr

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Olivier Godefroy

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Frederique Zindy

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Richard J. Gilbertson

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Daisuke Kawauchi

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Giles W. Robinson

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

David Finkelstein

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Jerold E. Rehg

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Amar Gajjar

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Carl W. Rettenmier

St. Jude Children's Research Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge