Network


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

Hotspot


Dive into the research topics where Marie McConkey is active.

Publication


Featured researches published by Marie McConkey.


Cell | 2011

Densely Interconnected Transcriptional Circuits Control Cell States in Human Hematopoiesis

Noa Novershtern; Aravind Subramanian; Lee N. Lawton; Raymond H. Mak; W. Nicholas Haining; Marie McConkey; Naomi Habib; Nir Yosef; Cindy Y. Chang; Tal Shay; Garrett M. Frampton; Adam Drake; Ilya B. Leskov; Björn Nilsson; Fred Preffer; David Dombkowski; John W. Evans; Ted Liefeld; John S. Smutko; Jianzhu Chen; Nir Friedman; Richard A. Young; Todd R. Golub; Aviv Regev; Benjamin L. Ebert

Though many individual transcription factors are known to regulate hematopoietic differentiation, major aspects of the global architecture of hematopoiesis remain unknown. Here, we profiled gene expression in 38 distinct purified populations of human hematopoietic cells and used probabilistic models of gene expression and analysis of cis-elements in gene promoters to decipher the general organization of their regulatory circuitry. We identified modules of highly coexpressed genes, some of which are restricted to a single lineage but most of which are expressed at variable levels across multiple lineages. We found densely interconnected cis-regulatory circuits and a large number of transcription factors that are differentially expressed across hematopoietic states. These findings suggest a more complex regulatory system for hematopoiesis than previously assumed.


Science | 2014

Lenalidomide Causes Selective Degradation of IKZF1 and IKZF3 in Multiple Myeloma Cells

Jan Krönke; Namrata D. Udeshi; Anupama Narla; Peter Grauman; Slater N. Hurst; Marie McConkey; Tanya Svinkina; Dirk Heckl; Eamon Comer; Xiaoyu Li; Christie Ciarlo; Emily Hartman; Nikhil C. Munshi; Monica Schenone; Stuart L. Schreiber; Steven A. Carr; Benjamin L. Ebert

Drug With a (Re)Purpose Thalidomide, once infamous for its deleterious effects on fetal development, has re-emerged as a drug of great interest because of its beneficial immunomodulatory effects. A derivative drug called lenalidomide significantly extends the survival of patients with multiple myeloma, but the molecular mechanisms underlying its efficacy remain unclear (see the Perspective by Stewart). Building on a previous observation that thalidomide binds to cereblon, a ubiquitin ligase, Lu et al. (p. 305, published online 28 November) and Krönke et al. (p. 301, published online 28 November) show that in the presence of lenalidomide, cereblon selectively targets two B cell transcription factors (Ikaros family members, IKZF1 and IKZF3) for degradation. In myeloma cell lines and patient cells, down-regulation of IKZF1 and IKZF3 was necessary and sufficient for the drugs anticancer activity. Thus, lenalidomide may act, at least in part, by “grepurposing” a ubiquitin ligase. A drug with potent activity in multiple myeloma patients acts by inducing degradation of two specific transcription factors. [Also see Perspective by Stewart] Lenalidomide is a drug with clinical efficacy in multiple myeloma and other B cell neoplasms, but its mechanism of action is unknown. Using quantitative proteomics, we found that lenalidomide causes selective ubiquitination and degradation of two lymphoid transcription factors, IKZF1 and IKZF3, by the CRBN-CRL4 ubiquitin ligase. IKZF1 and IKZF3 are essential transcription factors in multiple myeloma. A single amino acid substitution of IKZF3 conferred resistance to lenalidomide-induced degradation and rescued lenalidomide-induced inhibition of cell growth. Similarly, we found that lenalidomide-induced interleukin-2 production in T cells is due to depletion of IKZF1 and IKZF3. These findings reveal a previously unknown mechanism of action for a therapeutic agent: alteration of the activity of an E3 ubiquitin ligase, leading to selective degradation of specific targets.


Nature | 2015

Lenalidomide induces ubiquitination and degradation of CK1α in del(5q) MDS.

Jan Krönke; Emma C. Fink; Paul Hollenbach; Kyle J. MacBeth; Slater N. Hurst; Namrata D. Udeshi; Philip Chamberlain; D. R. Mani; Hon Wah Man; Anita Gandhi; Tanya Svinkina; Rebekka K. Schneider; Marie McConkey; Marcus Järås; Elizabeth A. Griffiths; Meir Wetzler; Lars Bullinger; Brian E. Cathers; Steven A. Carr; Rajesh Chopra; Benjamin L. Ebert

Lenalidomide is a highly effective treatment for myelodysplastic syndrome (MDS) with deletion of chromosome 5q (del(5q)). Here, we demonstrate that lenalidomide induces the ubiquitination of casein kinase 1A1 (CK1α) by the E3 ubiquitin ligase CUL4–RBX1–DDB1–CRBN (known as CRL4CRBN), resulting in CK1α degradation. CK1α is encoded by a gene within the common deleted region for del(5q) MDS and haploinsufficient expression sensitizes cells to lenalidomide therapy, providing a mechanistic basis for the therapeutic window of lenalidomide in del(5q) MDS. We found that mouse cells are resistant to lenalidomide but that changing a single amino acid in mouse Crbn to the corresponding human residue enables lenalidomide-dependent degradation of CK1α. We further demonstrate that minor side chain modifications in thalidomide and a novel analogue, CC-122, can modulate the spectrum of substrates targeted by CRL4CRBN. These findings have implications for the clinical activity of lenalidomide and related compounds, and demonstrate the therapeutic potential of novel modulators of E3 ubiquitin ligases.


The New England Journal of Medicine | 2017

Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease

Siddhartha Jaiswal; Pradeep Natarajan; Alexander J. Silver; Christopher J. Gibson; Alexander G. Bick; Eugenia Shvartz; Marie McConkey; Namrata Gupta; Stacey Gabriel; Diego Ardissino; Usman Baber; Roxana Mehran; Valentin Fuster; John Danesh; Philippe Frossard; Danish Saleheen; Olle Melander; Galina K. Sukhova; Donna Neuberg; Peter Libby; Sekar Kathiresan; Benjamin L. Ebert

BACKGROUND Clonal hematopoiesis of indeterminate potential (CHIP), which is defined as the presence of an expanded somatic blood‐cell clone in persons without other hematologic abnormalities, is common among older persons and is associated with an increased risk of hematologic cancer. We previously found preliminary evidence for an association between CHIP and atherosclerotic cardiovascular disease, but the nature of this association was unclear. METHODS We used whole‐exome sequencing to detect the presence of CHIP in peripheral‐blood cells and associated such presence with coronary heart disease using samples from four case–control studies that together enrolled 4726 participants with coronary heart disease and 3529 controls. To assess causality, we perturbed the function of Tet2, the second most commonly mutated gene linked to clonal hematopoiesis, in the hematopoietic cells of atherosclerosis‐prone mice. RESULTS In nested case–control analyses from two prospective cohorts, carriers of CHIP had a risk of coronary heart disease that was 1.9 times as great as in noncarriers (95% confidence interval [CI], 1.4 to 2.7). In two retrospective case–control cohorts for the evaluation of early‐onset myocardial infarction, participants with CHIP had a risk of myocardial infarction that was 4.0 times as great as in noncarriers (95% CI, 2.4 to 6.7). Mutations in DNMT3A, TET2, ASXL1, and JAK2 were each individually associated with coronary heart disease. CHIP carriers with these mutations also had increased coronary‐artery calcification, a marker of coronary atherosclerosis burden. Hypercholesterolemia‐prone mice that were engrafted with bone marrow obtained from homozygous or heterozygous Tet2 knockout mice had larger atherosclerotic lesions in the aortic root and aorta than did mice that had received control bone marrow. Analyses of macrophages from Tet2 knockout mice showed elevated expression of several chemokine and cytokine genes that contribute to atherosclerosis. CONCLUSIONS The presence of CHIP in peripheral‐blood cells was associated with nearly a doubling in the risk of coronary heart disease in humans and with accelerated atherosclerosis in mice. (Funded by the National Institutes of Health and others.)


Cancer Cell | 2014

Role of casein kinase 1A1 in the biology and targeted therapy of del(5q) MDS.

Rebekka K. Schneider; Vera Adema; Dirk Heckl; Marcus Järås; Mar Mallo; Allegra M. Lord; Lisa P. Chu; Marie McConkey; Rafael Kramann; Ann Mullally; Rafael Bejar; Francesc Solé; Benjamin L. Ebert

The casein kinase 1A1 gene (CSNK1A1) is a putative tumor suppressor gene located in the common deleted region for del(5q) myelodysplastic syndrome (MDS). We generated a murine model with conditional inactivation of Csnk1a1 and found that Csnk1a1 haploinsufficiency induces hematopoietic stem cell expansion and a competitive repopulation advantage, whereas homozygous deletion induces hematopoietic stem cell failure. Based on this finding, we found that heterozygous inactivation of Csnk1a1 sensitizes cells to a CSNK1 inhibitor relative to cells with two intact alleles. In addition, we identified recurrent somatic mutations in CSNK1A1 on the nondeleted allele of patients with del(5q) MDS. These studies demonstrate that CSNK1A1 plays a central role in the biology of del(5q) MDS and is a promising therapeutic target.


Blood | 2011

Dexamethasone and lenalidomide have distinct functional effects on erythropoiesis

Anupama Narla; Shilpee Dutt; J. Randall McAuley; Fatima Al-Shahrour; Slater N. Hurst; Marie McConkey; Donna Neuberg; Benjamin L. Ebert

Corticosteroids and lenalidomide decrease red blood cell transfusion dependence in patients with Diamond-Blackfan anemia (DBA) and myelodysplastic syndrome (MDS), respectively. We explored the effects of dexamethasone and lenalidomide, individually and in combination, on the differentiation of primary human bone marrow progenitor cells in vitro. Both agents promote erythropoiesis, increasing the absolute number of erythroid cells produced from normal CD34(+) cells and from CD34(+) cells with the types of ribosome dysfunction found in DBA and del(5q) MDS. However, the drugs had distinct effects on the production of erythroid progenitor colonies; dexamethasone selectively increased the number of burst-forming units-erythroid (BFU-E), whereas lenalidomide specifically increased colony-forming unit-erythroid (CFU-E). Use of the drugs in combination demonstrated that their effects are not redundant. In addition, dexamethasone and lenalidomide induced distinct gene-expression profiles. In coculture experiments, we examined the role of the microenvironment in response to both drugs and found that the presence of macrophages, the central cells in erythroblastic islands, accentuated the effects of both agents. Our findings indicate that dexamethasone and lenalidomide promote different stages of erythropoiesis and support the potential clinical utility of combination therapy for patients with bone marrow failure.


Cell | 2016

Core Circadian Clock Genes Regulate Leukemia Stem Cells in AML

Rishi V. Puram; Monika S. Kowalczyk; Carl G. de Boer; Rebekka K. Schneider; Peter Miller; Marie McConkey; Zuzana Tothova; Héctor Tejero; Dirk Heckl; Marcus Järås; Michelle Chen; Hubo Li; Alfred Tamayo; Glenn S. Cowley; Orit Rozenblatt-Rosen; Fatima Al-Shahrour; Aviv Regev; Benjamin L. Ebert

Leukemia stem cells (LSCs) have the capacity to self-renew and propagate disease upon serial transplantation in animal models, and elimination of this cell population is required for curative therapies. Here, we describe a series of pooled, in vivo RNAi screens to identify essential transcription factors (TFs) in a murine model of acute myeloid leukemia (AML) with genetically and phenotypically defined LSCs. These screens reveal the heterodimeric, circadian rhythm TFs Clock and Bmal1 as genes required for the growth of AML cells in vitro and in vivo. Disruption of canonical circadian pathway components produces anti-leukemic effects, including impaired proliferation, enhanced myeloid differentiation, and depletion of LSCs. We find that both normal and malignant hematopoietic cells harbor an intact clock with robust circadian oscillations, and genetic knockout models reveal a leukemia-specific dependence on the pathway. Our findings establish a role for the core circadian clock genes in AML.


Journal of Experimental Medicine | 2014

Csnk1a1 inhibition has p53-dependent therapeutic efficacy in acute myeloid leukemia

Marcus Järås; Peter Miller; Lisa P. Chu; Rishi V. Puram; Emma C. Fink; Rebekka K. Schneider; Fatima Al-Shahrour; Pablo Peña; L. Jordan Breyfogle; Kimberly A. Hartwell; Marie McConkey; Glenn S. Cowley; David E. Root; Michael G. Kharas; Ann Mullally; Benjamin L. Ebert

Targeting Csnk1a1 provides a potential therapeutic approach for AML associated with nonmutated Tp53.


Blood | 2015

EHMT1 and EHMT2 inhibition induces fetal hemoglobin expression

Aline Renneville; Matthew C. Canver; Marie McConkey; John M. Krill-Burger; David M. Dorfman; Edward B. Holson; Bradley E. Bernstein; Stuart H. Orkin; Daniel E. Bauer; Benjamin L. Ebert

Fetal hemoglobin (HbF, α2γ2) induction is a well-validated strategy for sickle cell disease (SCD) treatment. Using a small-molecule screen, we found that UNC0638, a selective inhibitor of EHMT1 and EHMT2 histone methyltransferases, induces γ-globin expression. EHMT1/2 catalyze mono- and dimethylation of lysine 9 on histone 3 (H3K9), raising the possibility that H3K9Me2, a repressive chromatin mark, plays a role in silencing γ-globin expression. In primary human adult erythroid cells, UNC0638 and EHMT1 or EHMT2 short hairpin RNA-mediated knockdown significantly increased γ-globin expression, HbF synthesis, and the percentage of cells expressing HbF. At effective concentrations, UNC0638 did not alter cell morphology, proliferation, or erythroid differentiation of primary human CD34(+) hematopoietic stem and progenitor cells in culture ex vivo. In murine erythroleukemia cells, UNC0638 and Ehmt2 CRISPR/Cas9-mediated knockout both led to a marked increase in expression of embryonic β-globin genes Hbb-εy and Hbb-βh1. In primary human adult erythroblasts, chromatin immunoprecipitation followed by sequencing analysis revealed that UNC0638 treatment leads to genome-wide depletion in H3K9Me2 and a concomitant increase in the activating mark H3K9Ac, which was especially pronounced at the γ-globin gene region. In RNA-sequencing analysis of erythroblasts, γ-globin genes were among the most significantly upregulated genes by UNC0638. Further increase in γ-globin expression in primary human adult erythroid cells was achieved by combining EHMT1/2 inhibition with the histone deacetylase inhibitor entinostat or hypomethylating agent decitabine. Our data provide genetic and pharmacologic evidence that EHMT1 and EHMT2 are epigenetic regulators involved in γ-globin repression and represent a novel therapeutic target for SCD.


Embo Molecular Medicine | 2017

Deletion of ribosomal protein genes is a common vulnerability in human cancer, especially in concert with TP53 mutations

Ram Ajore; David M. Raiser; Marie McConkey; Magnus Jöud; Bernd Boidol; Brenton G. Mar; Gordon Saksena; David M. Weinstock; Scott A. Armstrong; Steven R. Ellis; Benjamin L. Ebert; Björn Nilsson

Heterozygous inactivating mutations in ribosomal protein genes (RPGs) are associated with hematopoietic and developmental abnormalities, activation of p53, and altered risk of cancer in humans and model organisms. Here we performed a large‐scale analysis of cancer genome data to examine the frequency and selective pressure of RPG lesions across human cancers. We found that hemizygous RPG deletions are common, occurring in about 43% of 10,744 cancer specimens and cell lines. Consistent with p53‐dependent negative selection, such lesions are underrepresented in TP53‐intact tumors (P ≪ 10−10), and shRNA‐mediated knockdown of RPGs activated p53 in TP53‐wild‐type cells. In contrast, we did not see negative selection of RPG deletions in TP53‐mutant tumors. RPGs are conserved with respect to homozygous deletions, and shRNA screening data from 174 cell lines demonstrate that further suppression of hemizygously deleted RPGs inhibits cell growth. Our results establish RPG haploinsufficiency as a strikingly common vulnerability of human cancers that associates with TP53 mutations and could be targetable therapeutically.

Collaboration


Dive into the Marie McConkey's collaboration.

Top Co-Authors

Avatar

Benjamin L. Ebert

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ann Mullally

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Fatima Al-Shahrour

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rishi V. Puram

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Slater N. Hurst

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Dirk Heckl

Hannover Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge