Network


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

Hotspot


Dive into the research topics where Caroline Pabst is active.

Publication


Featured researches published by Caroline Pabst.


Cancer Cell | 2015

Inherited and Somatic Defects in DDX41 in Myeloid Neoplasms

Chantana Polprasert; Isabell Schulze; Mikkael A. Sekeres; Hideki Makishima; Bartlomiej Przychodzen; Naoko Hosono; Jarnail Singh; Richard A. Padgett; Xiaorong Gu; James G. Phillips; Michael J. Clemente; Yvonne Parker; Daniel J. Lindner; Brittney Dienes; Eckhard Jankowsky; Yogen Saunthararajah; Yang Du; Kevin Oakley; Nhu Nguyen; Sudipto Mukherjee; Caroline Pabst; Lucy A. Godley; Jane E. Churpek; Daniel A. Pollyea; Utz Krug; Wolfgang E. Berdel; Hans-Ulrich Klein; Martin Dugas; Yuichi Shiraishi; Kenichi Chiba

Most cases of adult myeloid neoplasms are routinely assumed to be sporadic. Here, we describe an adult familial acute myeloid leukemia (AML) syndrome caused by germline mutations in the DEAD/H-box helicase gene DDX41. DDX41 was also found to be affected by somatic mutations in sporadic cases of myeloid neoplasms as well as in a biallelic fashion in 50% of patients with germline DDX41 mutations. Moreover, corresponding deletions on 5q35.3 present in 6% of cases led to haploinsufficient DDX41 expression. DDX41 lesions caused altered pre-mRNA splicing and RNA processing. DDX41 is exemplary of other RNA helicase genes also affected by somatic mutations, suggesting that they constitute a family of tumor suppressor genes.


Genes & Development | 2014

The methyltransferase G9a regulates HoxA9-dependent transcription in AML

Bernhard Lehnertz; Caroline Pabst; Le Su; Michelle Miller; Feng Liu; Lin Yi; Regan Zhang; Jana Krosl; Eric Yung; Jeanette Kirschner; Patty Rosten; T. Michael Underhill; Jian Jin; Josée Hébert; Guy Sauvageau; R. Keith Humphries; Fabio Rossi

Chromatin modulators are emerging as attractive drug targets, given their widespread implication in human cancers and susceptibility to pharmacological inhibition. Here we establish the histone methyltransferase G9a/EHMT2 as a selective regulator of fast proliferating myeloid progenitors with no discernible function in hematopoietic stem cells (HSCs). In mouse models of acute myeloid leukemia (AML), loss of G9a significantly delays disease progression and reduces leukemia stem cell (LSC) frequency. We connect this function of G9a to its methyltransferase activity and its interaction with the leukemogenic transcription factor HoxA9 and provide evidence that primary human AML cells are sensitive to G9A inhibition. Our results highlight a clinical potential of G9A inhibition as a means to counteract the proliferation and self-renewal of AML cells by attenuating HoxA9-dependent transcription.


Clinical Cancer Research | 2007

The Graft Content of Donor T Cells Expressing γδTCR+ and CD4+foxp3+ Predicts the Risk of Acute Graft versus Host Disease after Transplantation of Allogeneic Peripheral Blood Stem Cells from Unrelated Donors

Caroline Pabst; Holger Schirutschke; Gerhard Ehninger; Martin Bornhäuser; Uwe Platzbecker

Purpose: Recently, high numbers of regulatory T cells within the stem cell graft were described to be associated with less graft-versus-host disease (GVHD) after related peripheral blood stem cell transplantation (PBSCT). Studies in mice also suggest a distinct role of γδTCR+ T cells in mediating GVHD. Therefore, the aim of this study was to define the yet-unknown role of regulatory and γδTCR+ T cells in human PBSCT from unrelated donors. Experimental Design: The frequency of both T-cell subsets within the graft was analyzed in 63 patients receiving unrelated allogeneic PBSCT. The respective amounts were quantified by flow cytometry and PCR and further correlated with clinical outcome. Results: The grafts contained a median of 11.2 × 106/kg CD4+foxp3+ and 9.8 × 106/kg γδTCR+ T cells, respectively. Patients receiving more CD4+foxp3+ cells had a lower cumulative incidence of acute GVHD II-IV (44% versus 65%, P = 0.03). Interestingly, in patients who received higher concentrations of donor γδTCR+ T cells, acute GVHD II-IV was more frequent (66% versus 40%, P = 0.02). In multivariate analysis, only the graft concentration of γδTCR+ T cells (P = 0.002) and a positive cytomegalovirus status of the recipient (P = 0.03) were significantly associated with the occurrence of acute GVHD II-IV. Conclusion: Graft composition of T-cell subsets seems to affect the outcome of patients receiving allogeneic PBSCT from unrelated donors. Therefore, selective manipulation or add-back of particular subsets might be a promising strategy to reduce the incidence of GVHD.


Nature Methods | 2014

Identification of small molecules that support human leukemia stem cell activity ex vivo

Caroline Pabst; Jana Krosl; Iman Fares; Geneviève Boucher; Réjean Ruel; Anne Marinier; Sébastien Lemieux; Josée Hébert; Guy Sauvageau

Leukemic stem cells (LSCs) are considered a major cause of relapse in acute myeloid leukemia (AML). Defining pathways that control LSC self-renewal is crucial for a better understanding of underlying mechanisms and for the development of targeted therapies. However, currently available culture conditions do not prevent spontaneous differentiation of LSCs, which greatly limits the feasibility of cell-based assays. To overcome these constraints we conducted a high-throughput chemical screen and identified small molecules that inhibit differentiation and support LSC activity in vitro. Similar to reports with cord blood stem cells, several of these compounds suppressed the aryl-hydrocarbon receptor (AhR) pathway, which we show to be inactive in vivo and rapidly activated ex vivo in AML cells. We also identified a compound, UM729, that collaborates with AhR suppressors in preventing AML cell differentiation. Together, these findings provide newly defined culture conditions for improved ex vivo culture of primary human AML cells.


Nature Medicine | 2017

Loss of the histone methyltransferase EZH2 induces resistance to multiple drugs in acute myeloid leukemia

Stefanie Göllner; Thomas Oellerich; Shuchi Agrawal-Singh; Tino Schenk; Hans-Ulrich Klein; Christian Rohde; Caroline Pabst; Tim Sauer; Mads Lerdrup; Sigal Tavor; Friedrich Stölzel; Sylvia Herold; Gerhard Ehninger; Gabriele Köhler; Kuan Ting Pan; Henning Urlaub; Hubert Serve; Martin Dugas; Karsten Spiekermann; Binje Vick; Irmela Jeremias; Wolfgang E. Berdel; Klaus Hansen; Arthur Zelent; Claudia Wickenhauser; Lutz P. Müller; Christian Thiede; Carsten Müller-Tidow

In acute myeloid leukemia (AML), therapy resistance frequently occurs, leading to high mortality among patients. However, the mechanisms that render leukemic cells drug resistant remain largely undefined. Here, we identified loss of the histone methyltransferase EZH2 and subsequent reduction of histone H3K27 trimethylation as a novel pathway of acquired resistance to tyrosine kinase inhibitors (TKIs) and cytotoxic drugs in AML. Low EZH2 protein levels correlated with poor prognosis in AML patients. Suppression of EZH2 protein expression induced chemoresistance of AML cell lines and primary cells in vitro and in vivo. Low EZH2 levels resulted in derepression of HOX genes, and knockdown of HOXB7 and HOXA9 in the resistant cells was sufficient to improve sensitivity to TKIs and cytotoxic drugs. The endogenous loss of EZH2 expression in resistant cells and primary blasts from a subset of relapsed AML patients resulted from enhanced CDK1-dependent phosphorylation of EZH2 at Thr487. This interaction was stabilized by heat shock protein 90 (HSP90) and followed by proteasomal degradation of EZH2 in drug-resistant cells. Accordingly, inhibitors of HSP90, CDK1 and the proteasome prevented EZH2 degradation, decreased HOX gene expression and restored drug sensitivity. Finally, patients with reduced EZH2 levels at progression to standard therapy responded to the combination of bortezomib and cytarabine, concomitant with the re-establishment of EZH2 expression and blast clearance. These data suggest restoration of EZH2 protein as a viable approach to overcome treatment resistance in this AML patient population.


Blood | 2016

GPR56 identifies primary human acute myeloid leukemia cells with high repopulating potential in vivo

Caroline Pabst; Anne Bergeron; Vincent-Philippe Lavallée; Jonathan Yeh; Patrick Gendron; Gudmundur L. Norddahl; Jana Krosl; Isabel Boivin; Eric Deneault; Jessica Simard; Suzan Imren; Geneviève Boucher; Kolja Eppert; Tobias Herold; Stefan K. Bohlander; Keith Humphries; Sébastien Lemieux; Josée Hébert; Guy Sauvageau; Frédéric Barabé

Acute myeloid leukemia (AML) is a genetically heterogeneous hematologic malignancy, which is initiated and driven by a rare fraction of leukemia stem cells (LSCs). Despite the difficulties of identifying a common LSC phenotype, there is increasing evidence that high expression of stem cell gene signatures is associated with poor clinical outcome. Identification of functionally distinct subpopulations in this disease is therefore crucial to dissecting the molecular machinery underlying LSC self-renewal. Here, we combined next-generation sequencing technology with in vivo assessment of LSC frequencies and identified the adhesion G protein-coupled receptor 56 (GPR56) as a novel and stable marker for human LSCs for the majority of AML samples. High GPR56 expression was significantly associated with high-risk genetic subgroups and poor outcome. Analysis of GPR56 in combination with CD34 expression revealed engraftment potential of GPR56(+)cells in both the CD34(-)and CD34(+)fractions, thus defining a novel LSC compartment independent of the CD34(+)CD38(-)LSC phenotype.


Blood | 2016

Chemo-genomic interrogation of CEBPA mutated AML reveals recurrent CSF3R mutations and subgroup sensitivity to JAK inhibitors.

Vincent-Philippe Lavallée; Jana Krosl; Sébastien Lemieux; Geneviève Boucher; Patrick Gendron; Caroline Pabst; Isabel Boivin; Anne Marinier; Cynthia J. Guidos; Sylvain Meloche; Josée Hébert; Guy Sauvageau

In this study, we analyzed RNA-sequencing data of 14 samples characterized by biallelic CEBPA (CEBPA(bi)) mutations included in the Leucegene collection of 415 primary acute myeloid leukemia (AML) specimens, and describe for the first time high frequency recurrent mutations in the granulocyte colony-stimulating factor receptor gene CSF3R, which signals through JAK-STAT proteins. Chemical interrogation of these primary human specimens revealed a uniform and specific sensitivity to all JAK inhibitors tested irrespective of their CSF3R mutation status, indicating a general sensitization of JAK-STAT signaling in this leukemia subset. Altogether, these results identified the co-occurrence of mutations in CSF3R and CEBPA in a well-defined AML subset, which uniformly responds to JAK inhibitors and paves the way to personalized clinical trials for this disease.


Blood Cancer Journal | 2016

Transcriptome analysis of G protein-coupled receptors in distinct genetic subgroups of acute myeloid leukemia: identification of potential disease-specific targets

Maiga A; Sébastien Lemieux; Caroline Pabst; Vincent-Philippe Lavallée; Michel Bouvier; Guy Sauvageau; Josée Hébert

Acute myeloid leukemia (AML) is associated with poor clinical outcome and the development of more effective therapies is urgently needed. G protein-coupled receptors (GPCRs) represent attractive therapeutic targets, accounting for approximately 30% of all targets of marketed drugs. Using next-generation sequencing, we studied the expression of 772 GPCRs in 148 genetically diverse AML specimens, normal blood and bone marrow cell populations as well as cord blood-derived CD34-positive cells. Among these receptors, 30 are overexpressed and 19 are downregulated in AML samples compared with normal CD34-positive cells. Upregulated GPCRs are enriched in chemokine (CCR1, CXCR4, CCR2, CX3CR1, CCR7 and CCRL2), adhesion (CD97, EMR1, EMR2 and GPR114) and purine (including P2RY2 and P2RY13) receptor subfamilies. The downregulated receptors include adhesion GPCRs, such as LPHN1, GPR125, GPR56, CELSR3 and GPR126, protease-activated receptors (F2R and F2RL1) and the Frizzled family receptors SMO and FZD6. Interestingly, specific deregulation was observed in genetically distinct subgroups of AML, thereby identifying different potential therapeutic targets in these frequent AML subgroups.


Blood | 2014

UBAP2L is a novel BMI1-interacting protein essential for hematopoietic stem cell activity

Marie-Eve Bordeleau; Romain Aucagne; Jalila Chagraoui; Simon Girard; Nadine Mayotte; Eric Bonneil; Pierre Thibault; Caroline Pabst; Anne Bergeron; Frédéric Barabé; Josée Hébert; Martin Sauvageau; Christel Boutonnet; Sylvain Meloche; Guy Sauvageau

Multipotent long-term repopulating hematopoietic stem cells (LT-HSCs) can self-renew or differentiate into the less primitive short-term repopulating stem cells (ST-HSCs), which themselves produce progenitors that ensure the daily supply of all essential blood components. The Polycomb group (PcG) protein BMI1 is essential for the activity of both HSCs and progenitor cells. Although BMI1 operates by suppressing the Ink4a/Arf locus in progenitors and ST-HSCs, the mechanisms through which this gene regulates the activity of LT-HSCs remain poorly understood. Toward this goal, we isolated BMI1-containing protein complexes and identified UBAP2L as a novel BMI1-interacting protein. We also showed that UBAP2L is preferentially expressed in mouse and human HSC-enriched populations when compared with more mature cell types, and that this gene is essential for the activity of LT-HSCs. In contrast to what is observed for Bmi1 knockdown, we found that UBAP2L depletion does not affect the Ink4a/Arf locus. Given that we demonstrated that BMI1 overexpression is able to rescue the deleterious effects of Ubap2l downregulation on LT-HSC activity and that UBAP2L is part of a PcG subcomplex comprising BMI1, we propose a model in which at least 2 different BMI1-containing PcG complexes regulate HSC activity, which are distinguishable by the presence of UBAP2L.


Advances in Immunology | 2017

Adhesion GPCRs in Regulating Immune Responses and Inflammation

Hsi-Hsien Lin; Cheng-Chih Hsiao; Caroline Pabst; Josée Hébert; Torsten Schöneberg; Jörg Hamann

The adhesion family comprises one of the five major clades of G protein-coupled receptors (GPCRs). Unlike conventional GPCRs, adhesion GPCRs (aGPCRs) have extended ectodomains with various protein folds that facilitate protein-protein interactions and, hence, putative cellular adhesive functions. Juxtaposed to the seven-pass transmembrane domain is a GPCR autoproteolysis-inducing domain that enables autoproteolytic cleavage of the receptor, resulting in a bipartite structure of many aGPCRs. aGPCRs are widely distributed and play critical roles in many developmental processes; yet, the underlying mechanisms of activation and signal transduction have emerged only recently. About one-third of the 33 human aGPCRs are expressed in hematopoietic stem, progenitor, or mature cells, where they define distinct cellular populations. Recent studies have demonstrated roles of aGPCR in the control of innate effector functions and the susceptibility for and onset of (auto)inflammatory conditions. We here discuss the current knowledge about aGPCRs in the regulation of immune responses and inflammation.

Collaboration


Dive into the Caroline Pabst's collaboration.

Top Co-Authors

Avatar

Guy Sauvageau

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar

Josée Hébert

Hôpital Maisonneuve-Rosemont

View shared research outputs
Top Co-Authors

Avatar

Jana Krosl

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gerhard Ehninger

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Martin Bornhäuser

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Uwe Platzbecker

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge