Network


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

Hotspot


Dive into the research topics where Marion Lebois is active.

Publication


Featured researches published by Marion Lebois.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Mpl expression on megakaryocytes and platelets is dispensable for thrombopoiesis but essential to prevent myeloproliferation

Ashley P. Ng; Maria Kauppi; Donald Metcalf; Craig D. Hyland; Emma C. Josefsson; Marion Lebois; Jian-Guo Zhang; Tracey M. Baldwin; Ladina Di Rago; Douglas J. Hilton; Warren S. Alexander

Significance Blood platelets, the small circulating cells that coordinate hemostasis, are produced by specialized bone marrow cells called megakaryocytes. The cytokine thrombopoietin (TPO) is a key regulator of platelet production acting via its specific cell receptor, Mpl. Via genetic modification of the Mpl allele in mice, we precisely define the bone marrow cells that express Mpl and, by genetically removing Mpl from megakaryocytes and platelets, we show TPO signaling via Mpl is not required in megakaryocytes for their expansion, maturation, or platelet production. Rather, Mpl expression on megakaryocytes is essential for regulating TPO availability in the bone marrow microenvironment to prevent myeloproliferation, a model we suggest is important for human disease. Thrombopoietin (TPO) acting via its receptor, the cellular homologue of the myeloproliferative leukemia virus oncogene (Mpl), is the major cytokine regulator of platelet number. To precisely define the role of specific hematopoietic cells in TPO-dependent hematopoiesis, we generated mice that express the Mpl receptor normally on stem/progenitor cells but lack expression on megakaryocytes and platelets (MplPF4cre/PF4cre). MplPF4cre/PF4cre mice displayed profound megakaryocytosis and thrombocytosis with a remarkable expansion of megakaryocyte-committed and multipotential progenitor cells, the latter displaying biological responses and a gene expression signature indicative of chronic TPO overstimulation as the underlying causative mechanism, despite a normal circulating TPO level. Thus, TPO signaling in megakaryocytes is dispensable for platelet production; its key role in control of platelet number is via generation and stimulation of the bipotential megakaryocyte precursors. Nevertheless, Mpl expression on megakaryocytes and platelets is essential to prevent megakaryocytosis and myeloproliferation by restricting the amount of TPO available to stimulate the production of megakaryocytes from the progenitor cell pool.


Blood | 2012

Mcl-1 and Bcl-xL coordinately regulate megakaryocyte survival

Marlyse A. Debrincat; Emma C. Josefsson; Chloé James; Katya J. Henley; Sarah Ellis; Marion Lebois; Kelly L. Betterman; Rachael M. Lane; Kelly L. Rogers; M. J. D. White; Andrew W. Roberts; Natasha L. Harvey; Donald Metcalf; Benjamin T. Kile

Mature megakaryocytes depend on the function of Bcl-x(L), a member of the Bcl-2 family of prosurvival proteins, to proceed safely through the process of platelet shedding. Despite this, loss of Bcl-x(L) does not prevent the growth and maturation of megakaryocytes, suggesting redundancy with other prosurvival proteins. We therefore generated mice with a megakaryocyte-specific deletion of Mcl-1, which is known to be expressed in megakaryocytes. Megakaryopoiesis, platelet production, and platelet lifespan were unperturbed in Mcl-1(Pf4Δ/Pf4Δ) animals. However, treatment with ABT-737, a BH3 mimetic compound that inhibits the prosurvival proteins Bcl-2, Bcl-x(L), and Bcl-w resulted in the complete ablation of megakaryocytes and platelets. Genetic deletion of both Mcl-1 and Bcl-x(L) in megakaryocytes resulted in preweaning lethality. Megakaryopoiesis in Bcl-x(Pf4Δ/Pf4Δ) Mcl-1(Pf4Δ/Pf4Δ) embryos was severely compromised, and these animals exhibited ectopic bleeding. Our studies indicate that the combination of Bcl-x(L) and Mcl-1 is essential for the viability of the megakaryocyte lineage.


Blood | 2012

Mcl-1 and Bcl-xL co-ordinately regulate megakaryocyte survival

Marlyse A. Debrincat; Emma C. Josefsson; Chloé James; Katya J. Henley; Sarah Ellis; Marion Lebois; Kelly L. Betterman; Rachael M. Lane; Kelly L. Rogers; M. J. D. White; Andrew W. Roberts; Natasha L. Harvey; Donald Metcalf; Benjamin T. Kile

Mature megakaryocytes depend on the function of Bcl-x(L), a member of the Bcl-2 family of prosurvival proteins, to proceed safely through the process of platelet shedding. Despite this, loss of Bcl-x(L) does not prevent the growth and maturation of megakaryocytes, suggesting redundancy with other prosurvival proteins. We therefore generated mice with a megakaryocyte-specific deletion of Mcl-1, which is known to be expressed in megakaryocytes. Megakaryopoiesis, platelet production, and platelet lifespan were unperturbed in Mcl-1(Pf4Δ/Pf4Δ) animals. However, treatment with ABT-737, a BH3 mimetic compound that inhibits the prosurvival proteins Bcl-2, Bcl-x(L), and Bcl-w resulted in the complete ablation of megakaryocytes and platelets. Genetic deletion of both Mcl-1 and Bcl-x(L) in megakaryocytes resulted in preweaning lethality. Megakaryopoiesis in Bcl-x(Pf4Δ/Pf4Δ) Mcl-1(Pf4Δ/Pf4Δ) embryos was severely compromised, and these animals exhibited ectopic bleeding. Our studies indicate that the combination of Bcl-x(L) and Mcl-1 is essential for the viability of the megakaryocyte lineage.


Nature Communications | 2014

Platelet production proceeds independently of the intrinsic and extrinsic apoptosis pathways

Emma C. Josefsson; Deborah L. Burnett; Marion Lebois; Marlyse A. Debrincat; Michael J. White; Katya J. Henley; Rachael M. Lane; Diane Moujalled; Simon Preston; Lorraine A. O'Reilly; Marc Pellegrini; Donald Metcalf; Andreas Strasser; Benjamin T. Kile

BH3 mimetic drugs that target BCL-2 family pro-survival proteins to induce tumour cell apoptosis represent a new era in cancer therapy. Clinical trials of navitoclax (ABT-263, which targets BCL-2, BCL-XL and BCL-W) have shown great promise, but encountered dose-limiting thrombocytopenia. Recent work has demonstrated that this is due to the inhibition of BCL-XL, which is essential for platelet survival. These findings raise new questions about the established model of platelet shedding by megakaryocytes, which is thought to be an apoptotic process. Here we generate mice with megakaryocyte-specific deletions of the essential mediators of extrinsic (Caspase-8) and intrinsic (BAK/BAX) apoptosis. We show that megakaryocytes possess a Fas ligand-inducible extrinsic apoptosis pathway. However, Fas activation does not stimulate platelet production, rather, it triggers Caspase-8-mediated killing. Combined loss of Caspase-8/BAK/BAX does not impair thrombopoiesis, but can protect megakaryocytes from death in mice infected with lymphocytic choriomeningitis virus. Thus, apoptosis is dispensable for platelet biogenesis.


Blood | 2015

Polycomb repressive complex 2 component Suz12 is required for hematopoietic stem cell function and lymphopoiesis.

Stanley Chun-Wei Lee; Sarah E. Miller; Craig D. Hyland; Maria Kauppi; Marion Lebois; Ladina Di Rago; Donald Metcalf; Sarah Kinkel; Emma C. Josefsson; Marnie E. Blewitt; Ian Majewski; Warren S. Alexander

Polycomb repressive complex 2 (PRC2) is a chromatin modifier that regulates stem cells in embryonic and adult tissues. Loss-of-function studies of PRC2 components have been complicated by early embryonic dependence on PRC2 activity and the partial functional redundancy of enhancer of zeste homolog 1 (Ezh1) and enhancer of zeste homolog 2 (Ezh2), which encode the enzymatic component of PRC2. Here, we investigated the role of PRC2 in hematopoiesis by conditional deletion of suppressor of zeste 12 protein homolog (Suz12), a core component of PRC2. Complete loss of Suz12 resulted in failure of hematopoiesis, both in the embryo and the adult, with a loss of maintenance of hematopoietic stem cells (HSCs). In contrast, partial loss of PRC2 enhanced HSC self-renewal. Although Suz12 was required for lymphoid development, deletion in individual blood cell lineages revealed that it was dispensable for the development of granulocytic, monocytic, and megakaryocytic cells. Collectively, these data reveal the multifaceted role of PRC2 in hematopoiesis, with divergent dose-dependent effects in HSC and distinct roles in maturing blood cells. Because PRC2 is a potential target for cancer therapy, the significant consequences of modest changes in PRC2 activity, as well as the cell and developmental stage-specific effects, will need to be carefully considered in any therapeutic context.


Cell Death and Disease | 2015

BCL-2 is dispensable for thrombopoiesis and platelet survival

Marlyse A. Debrincat; Irina Pleines; Marion Lebois; Rachael M. Lane; Melissa L. Holmes; Jason Corbin; Cassandra J. Vandenberg; Warren S. Alexander; Ashley P. Ng; Andreas Strasser; Martha Sola-Visner; Benjamin T. Kile; Emma C. Josefsson

Navitoclax (ABT-263), an inhibitor of the pro-survival BCL-2 family proteins BCL-2, BCL-XL and BCL-W, has shown clinical efficacy in certain BCL-2-dependent haematological cancers, but causes dose-limiting thrombocytopaenia. The latter effect is caused by Navitoclax directly inducing the apoptotic death of platelets, which are dependent on BCL-XL for survival. Recently, ABT-199, a selective BCL-2 antagonist, was developed. It has shown promising anti-leukaemia activity in patients whilst sparing platelets, suggesting that the megakaryocyte lineage does not require BCL-2. In order to elucidate the role of BCL-2 in megakaryocyte and platelet survival, we generated mice with a lineage-specific deletion of Bcl2, alone or in combination with loss of Mcl1 or Bclx. Platelet production and platelet survival were analysed. Additionally, we made use of BH3 mimetics that selectively inhibit BCL-2 or BCL-XL. We show that the deletion of BCL-2, on its own or in concert with MCL-1, does not affect platelet production or platelet lifespan. Thrombocytopaenia in Bclx-deficient mice was not affected by additional genetic loss or pharmacological inhibition of BCL-2. Thus, BCL-2 is dispensable for thrombopoiesis and platelet survival in mice.


Platelets | 2016

Regulation of platelet lifespan by apoptosis

Marion Lebois; Emma C. Josefsson

Abstract The lifespan of platelets in circulation is brief, close to 10 days in humans and 5 days in mice. Bone marrow residing megakaryocytes produce around 100 billion platelets per day. In a healthy individual, the majority of platelets are not consumed by hemostatic processes, but rather their lifespan is controlled by programmed cell death, a canonical intrinsic apoptosis program. In the last decade, insights from genetically manipulated mouse models and pharmacological developments have helped to define the components of the intrinsic, or mitochondrial, apoptosis pathway that controls platelet lifespan. This review focuses on the molecular regulation of apoptosis in platelet survival, reviews thrombocytopenic conditions linked to enhanced platelet death, examines implications of chemotherapy-induced thrombocytopenia through apoptosis-inducing drugs in cancer therapy as well as discusses ex vivo aging of platelets.


Platelets (Third Edition) | 2013

The Regulation of Platelet Life Span

Emma C. Josefsson; Mark R. Dowling; Marion Lebois; Benjamin T. Kile

The human body produces approximately 100 billion platelets per day. At steady state, only a small fraction of these are consumed in hemostatic processes. The majority are cleared by the reticuloendothelial system in the liver and spleen. Their time in the circulation is relatively brief; up to 10 days in humans and 5 days in mice. In recent years it has become apparent that platelets contain an apoptotic program that regulates their survival. Genetic or pharmacological manipulation of the program can reduce or extend platelet life span in vivo . This chapter reviews the history of platelet life span research, highlights recent developments, and outlines some emerging themes in the field.


Journal of Thrombosis and Haemostasis | 2016

Regulation of platelet lifespan in the presence and absence of thrombopoietin signaling.

Marion Lebois; Mark R. Dowling; Pradnya Gangatirkar; Philip D. Hodgkin; Benjamin T. Kile; Warren S. Alexander; Emma C. Josefsson

Essentials We examined platelet survival in models of absent or enhanced thrombopoietin (TPO) signaling. Platelet lifespan is normal in transgenic mice with chronically enhanced TPO signaling. Mpl deficiency does not negatively affect platelet lifespan in the absence of thrombocytopenia. We conclude that TPO and its receptor Mpl are dispensable for platelet survival in adult mice.


Blood | 2018

Intrinsic apoptosis circumvents the functional decline of circulating platelets but does not cause the storage lesion

Irina Pleines; Marion Lebois; Pradnya Gangatirkar; Amanda E. Au; Rachael M. Lane; Katya J. Henley; Maria Kauppi; Jason Corbin; Ping Cannon; Jonathan P. Bernardini; Imala Alwis; Kate E. Jarman; Sarah Ellis; Donald Metcalf; Shaun P. Jackson; Simone M. Schoenwaelder; Benjamin T. Kile; Emma C. Josefsson

The circulating life span of blood platelets is regulated by the prosurvival protein BCL-XL It restrains the activity of BAK and BAX, the essential prodeath mediators of intrinsic apoptosis. Disabling the platelet intrinsic apoptotic pathway in mice by deleting BAK and BAX results in a doubling of platelet life span and concomitant thrombocytosis. Apoptotic platelets expose phosphatidylserine (PS) via a mechanism that is distinct from that driven by classical agonists. Whether there is any role for apoptotic PS in platelet function in vivo, however, is unclear. Apoptosis has also been associated with the platelet storage lesion (PSL), the constellation of biochemical deteriorations that occur during blood bank storage. In this study, we investigated the role of BAK/BAX-mediated apoptosis in hemostasis and thrombosis and in the development of the PSL. We show that although intrinsic apoptosis is rapidly induced during storage at 37°C, it is not detected when platelets are kept at the standard storage temperature of 22°C. Remarkably, loss of BAK and BAX did not prevent the development of the PSL at either temperature. BAK/BAX-deficient mice exhibited increased bleeding times and unstable thrombus formation. This phenotype was not caused by impaired PS exposure, but was associated with a defect in granule release from aged platelets. Strikingly, rejuvenation of BAK/BAX-deficient platelets in vivo completely rescued the observed hemostatic defects. Thus, apoptotic culling of old platelets from the bloodstream is essential to maintain a functional, hemostatically reactive platelet population. Inhibiting intrinsic apoptosis in blood banked platelets is unlikely to yield significant benefit.

Collaboration


Dive into the Marion Lebois's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Benjamin T. Kile

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Donald Metcalf

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Rachael M. Lane

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Warren S. Alexander

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Katya J. Henley

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Marlyse A. Debrincat

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andreas Strasser

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Jason Corbin

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge