Network


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

Hotspot


Dive into the research topics where Miriam Sansó is active.

Publication


Featured researches published by Miriam Sansó.


Nature Structural & Molecular Biology | 2012

Cyclin-dependent kinase control of the initiation-to-elongation switch of RNA polymerase II

Stéphane Larochelle; Ramon Amat; Kira Glover-Cutter; Miriam Sansó; Chao Zhang; Jasmina J. Allen; Kevan M. Shokat; David L. Bentley; Robert P. Fisher

Promoter-proximal pausing by RNA polymerase II (Pol II) ensures gene-specific regulation and RNA quality control. Structural considerations suggested a requirement for initiation-factor eviction in elongation-factor engagement and pausing of transcription complexes. Here we show that selective inhibition of Cdk7—part of TFIIH—increases TFIIE retention, prevents DRB sensitivity–inducing factor (DSIF) recruitment and attenuates pausing in human cells. Pause release depends on Cdk9–cyclin T1 (P-TEFb); Cdk7 is also required for Cdk9-activating phosphorylation and Cdk9-dependent downstream events—Pol II C-terminal domain Ser2 phosphorylation and histone H2B ubiquitylation—in vivo. Cdk7 inhibition, moreover, impairs Pol II transcript 3′-end formation. Cdk7 thus acts through TFIIE and DSIF to establish, and through P-TEFb to relieve, barriers to elongation: incoherent feedforward that might create a window to recruit RNA-processing machinery. Therefore, cyclin-dependent kinases govern Pol II handoff from initiation to elongation factors and cotranscriptional RNA maturation.


PLOS Genetics | 2012

A positive feedback loop links opposing functions of P-TEFb/Cdk9 and histone H2B ubiquitylation to regulate transcript elongation in fission yeast.

Miriam Sansó; Karen Shi Mei Lee; Laia Viladevall; Pierre-Étienne Jacques; Viviane Pagé; Stephen Nagy; Ariane Racine; Courtney V. St. Amour; Chao Zhang; Kevan M. Shokat; Beate Schwer; François Robert; Robert P. Fisher; Jason C. Tanny

Transcript elongation by RNA polymerase II (RNAPII) is accompanied by conserved patterns of histone modification. Whereas histone modifications have established roles in transcription initiation, their functions during elongation are not understood. Mono-ubiquitylation of histone H2B (H2Bub1) plays a key role in coordinating co-transcriptional histone modification by promoting site-specific methylation of histone H3. H2Bub1 also regulates gene expression through an unidentified, methylation-independent mechanism. Here we reveal bidirectional communication between H2Bub1 and Cdk9, the ortholog of metazoan positive transcription elongation factor b (P-TEFb), in the fission yeast Schizosaccharomyces pombe. Chemical and classical genetic analyses indicate that lowering Cdk9 activity or preventing phosphorylation of its substrate, the transcription processivity factor Spt5, reduces H2Bub1 in vivo. Conversely, mutations in the H2Bub1 pathway impair Cdk9 recruitment to chromatin and decrease Spt5 phosphorylation. Moreover, an Spt5 phosphorylation-site mutation, combined with deletion of the histone H3 Lys4 methyltransferase Set1, phenocopies morphologic and growth defects due to H2Bub1 loss, suggesting independent, partially redundant roles for Cdk9 and Set1 downstream of H2Bub1. Surprisingly, mutation of the histone H2B ubiquitin-acceptor residue relaxes the Cdk9 activity requirement in vivo, and cdk9 mutations suppress cell-morphology defects in H2Bub1-deficient strains. Genome-wide analyses by chromatin immunoprecipitation also demonstrate opposing effects of Cdk9 and H2Bub1 on distribution of transcribing RNAPII. Therefore, whereas mutual dependence of H2Bub1 and Spt5 phosphorylation indicates positive feedback, mutual suppression by cdk9 and H2Bub1-pathway mutations suggests antagonistic functions that must be kept in balance to regulate elongation. Loss of H2Bub1 disrupts that balance and leads to deranged gene expression and aberrant cell morphologies, revealing a novel function of a conserved, co-transcriptional histone modification.


Molecular and Cellular Biology | 2012

Separate Domains of Fission Yeast Cdk9 (P-TEFb) Are Required for Capping Enzyme Recruitment and Primed (Ser7-Phosphorylated) Rpb1 Carboxyl-Terminal Domain Substrate Recognition

Courtney V. St. Amour; Miriam Sansó; Christian A. Bösken; Karen M. Lee; Stéphane Larochelle; Chao Zhang; Kevan M. Shokat; Matthias Geyer; Robert P. Fisher

ABSTRACT In fission yeast, discrete steps in mRNA maturation and synthesis depend on a complex containing the 5′-cap methyltransferase Pcm1 and Cdk9, which phosphorylates the RNA polymerase II (Pol II) carboxyl-terminal domain (CTD) and the processivity factor Spt5 to promote transcript elongation. Here we show that a Cdk9 carboxyl-terminal extension, distinct from the catalytic domain, mediates binding to both Pcm1 and the Pol II CTD. Removal of this segment diminishes Cdk9/Pcm1 chromatin recruitment and Spt5 phosphorylation in vivo and leads to slow growth and hypersensitivity to cold temperature, nutrient limitation, and the IMP dehydrogenase inhibitor mycophenolic acid (MPA). These phenotypes, and the Spt5 phosphorylation defect, are suppressed by Pcm1 overproduction, suggesting that normal transcript elongation and gene expression depend on physical linkage between Cdk9 and Pcm1. The extension is dispensable, however, for recognition of CTD substrates “primed” by Mcs6 (Cdk7). On defined peptide substrates in vitro, Cdk9 prefers CTD repeats phosphorylated at Ser7 over unmodified repeats. In vivo, Ser7 phosphorylation depends on Mcs6 activity, suggesting a conserved mechanism, independent of chromatin recruitment, to order transcriptional CDK functions. Therefore, fission yeast Cdk9 comprises a catalytic domain sufficient for primed substrate recognition and a multivalent recruitment module that couples transcription with capping.


Transcription | 2013

Pause, play, repeat: CDKs push RNAP II's buttons.

Miriam Sansó; Robert P. Fisher

Cyclin-dependent kinases (CDKs) play a central role in governing eukaryotic cell division. It is becoming clear that the transcription cycle of RNA polymerase II (RNAP II) is also regulated by CDKs; in metazoans, the cell cycle and transcriptional CDK networks even share an upstream activating kinase, which is itself a CDK. From recent chemical-genetic analyses we know that CDKs and their substrates control events both early in transcription (the transition from initiation to elongation) and late (3′ end formation and transcription termination). Moreover, mutual dependence on CDK activity might couple the “beginning” and “end” of the cycle, to ensure the fidelity of mRNA maturation and the efficient recycling of RNAP II from sites of termination to the transcription start site (TSS). As is the case for CDKs involved in cell cycle regulation, different transcriptional CDKs act in defined sequence on multiple substrates. These phosphorylations are likely to influence gene expression by several mechanisms, including direct, allosteric effects on the transcription machinery, co-transcriptional recruitment of proteins needed for mRNA-capping, splicing and 3′ end maturation, dependent on multisite phosphorylation of the RNAP II C-terminal domain (CTD) and, perhaps, direct regulation of RNA-processing or histone-modifying machinery. Here we review these recent advances, and preview the emerging challenges for transcription-cycle research.


Journal of Clinical Investigation | 2017

Neonatal expression of RNA-binding protein IGF2BP3 regulates the human fetal-adult megakaryocyte transition

Kamaleldin E. Elagib; Chih Huan Lu; Goar Mosoyan; Shadi Khalil; Ewelina Zasadzińska; Daniel R. Foltz; Peter Balogh; Alejandro A. Gru; Deborah A. Fuchs; Lisa M. Rimsza; Els Verhoeyen; Miriam Sansó; Robert P. Fisher; Camelia Iancu-Rubin; Adam N. Goldfarb

Hematopoietic transitions that accompany fetal development, such as erythroid globin chain switching, play important roles in normal physiology and disease development. In the megakaryocyte lineage, human fetal progenitors do not execute the adult morphogenesis program of enlargement, polyploidization, and proplatelet formation. Although these defects decline with gestational stage, they remain sufficiently severe at birth to predispose newborns to thrombocytopenia. These defects may also contribute to inferior platelet recovery after cord blood stem cell transplantation and may underlie inefficient platelet production by megakaryocytes derived from pluripotent stem cells. In this study, comparison of neonatal versus adult human progenitors has identified a blockade in the specialized positive transcription elongation factor b (P-TEFb) activation mechanism that is known to drive adult megakaryocyte morphogenesis. This blockade resulted from neonatal-specific expression of an oncofetal RNA-binding protein, IGF2BP3, which prevented the destabilization of the nuclear RNA 7SK, a process normally associated with adult megakaryocytic P-TEFb activation. Knockdown of IGF2BP3 sufficed to confer both phenotypic and molecular features of adult-type cells on neonatal megakaryocytes. Pharmacologic inhibition of IGF2BP3 expression via bromodomain and extraterminal domain (BET) inhibition also elicited adult features in neonatal megakaryocytes. These results identify IGF2BP3 as a human ontogenic master switch that restricts megakaryocyte development by modulating a lineage-specific P-TEFb activation mechanism, revealing potential strategies toward enhancing platelet production.


Biochemical Society Transactions | 2013

Modelling the CDK-dependent transcription cycle in fission yeast

Miriam Sansó; Robert P. Fisher

CDKs (cyclin-dependent kinases) ensure directionality and fidelity of the eukaryotic cell division cycle. In a similar fashion, the transcription cycle is governed by a conserved subfamily of CDKs that phosphorylate Pol II (RNA polymerase II) and other substrates. A genetic model organism, the fission yeast Schizosaccharomyces pombe, has yielded robust models of cell-cycle control, applicable to higher eukaryotes. From a similar approach combining classical and chemical genetics, fundamental principles of transcriptional regulation by CDKs are now emerging. In the present paper, we review the current knowledge of each transcriptional CDK with respect to its substrate specificity, function in transcription and effects on chromatin modifications, highlighting the important roles of CDKs in ensuring quantity and quality control over gene expression in eukaryotes.


bioRxiv | 2017

A Cdk9-PP1 kinase-phosphatase switch regulates the elongation-termination transition of RNA polymerase II

Pabitra K Parua; Gregory T. Booth; Miriam Sansó; Bradley Benjamin; Jason C. Tanny; John T. Lis; Robert P. Fisher

The end of the RNA polymerase II (Pol II) transcription cycle is strictly regulated to ensure proper mRNA maturation and prevent interference between neighboring genes1. Pol II slowing downstream of the cleavage and polyadenylation signal (CPS) leads to recruitment of cleavage and polyadenylation factors and termination2, but how this chain of events is initiated remains unclear. In a chemical-genetic screen, we identified protein phosphatase 1 (PP1) isoforms as substrates of human positive transcription elongation factor b (P-TEFb), the cyclin-dependent kinase 9 (Cdk9)-cyclin T1 complex3. Here we show that Cdk9 and PP1 govern phosphorylation of the conserved transcription factor Spt5 in the fission yeast Schizosaccharomyces pombe. Cdk9 phosphorylates both Spt5 and a negative regulatory site on the PP1 isoform Dis24. Sites phosphorylated by Cdk9 in the Spt5 carboxy-terminal domain (CTD) are dephosphorylated by Dis2 in vitro, and Cdk9 inhibition in vivo leads to rapid Spt5 dephosphorylation that is retarded by concurrent Dis2 inactivation. Chromatin immunoprecipitation and sequencing (ChIP-seq) analysis indicates that Spt5 is dephosphorylated as transcription complexes traverse the CPS, prior to or concomitant with slowing of Pol II5. A Dis2-inactivating mutation stabilizes Spt5 phosphorylation (pSpt5) on chromatin, promotes transcription beyond the normal termination zone detected by precision run-on transcription and sequencing (PRO-seq)6, and is suppressed by ablation of Cdk9 target sites in Spt5. These results support a model whereby the transition of Pol II from elongation to termination is regulated by opposing activities of Cdk9 and Dis2 towards their common substrate Spt5—a bistable switch analogous to a Cdk1-PP1 module that controls mitotic progression4.


bioRxiv | 2017

Cdk9 regulates a promoter-proximal checkpoint to modulate RNA Polymerase II elongation rate

Gregory T. Booth; Pabitra K Parua; Miriam Sansó; Robert P. Fisher; John T. Lis

Multiple kinases modify RNA Polymerase II (Pol II) and its associated pausing and elongation factors to regulate Pol II transcription and transcription-coupled mRNA processing1,2. The conserved Cdk9 kinase is essential for regulated eukaryotic transcription3, but its mechanistic role remains incompletely understood. Here, we use altered-specificity kinase mutations and highly-specific inhibitors in fission yeast, Schizosaccharomyces pombe to examine the role of Cdk9, and related Cdk7 and Cdk12 kinases, on transcription at base-pair resolution using Precision Run-On sequencing (PRO-seq). Within a minute, Cdk9 inhibition causes a dramatic reduction in the phosphorylation of Pol II-associated factor, Spt5. The effects of Cdk9 inhibition on transcription are the more severe than inhibition of Cdk7 and Cdk12 and result in a shift of Pol II towards the transcription start site (TSS). A kinetic time course of Cdk9 inhibition reveals that early transcribing Pol II is the most compromised, with a measured rate of only ~400 bp/min, while Pol II that is already well into the gene continues rapidly to the end of genes with a rate > 1 kb/min. Our results indicate that while Pol II in S. pombe can escape promoter-proximal pausing in the absence of Cdk9 activity, it is impaired in elongation, suggesting the existence of a conserved global regulatory checkpoint that requires Cdk9 kinase activity.


bioRxiv | 2017

Cdk9, Spt5 and histone H2B mono-ubiquitylation cooperate to ensure antisense suppression by the Clr6-CII/Rpd3S HDAC complex

Miriam Sansó; Daniel Pinto; Peter Svensson; Viviane Pagé; Pabitra K Parua; Danny A. Bitton; Jean Mbogning; Patricia García; Elena Hidalgo; François Robert; Jürg Bähler; Jason C. Tanny; Robert P. Fisher

Cyclin-dependent kinase 9 (Cdk9) and histone H2B monoubiquitylation (H2Bub1) are both implicated in elongation by RNA polymerase II (RNAPII). In fission yeast, Cdk9 and H2Bub1 regulate each other through a feedback loop involving phosphorylation of the elongation factor Spt5. Conversely, genetic interactions suggest opposing functions of H2Bub1 and Cdk9 through an Spt5-independent pathway. To understand these interactions, we performed RNA-seq analysis after H2Bub1 loss, Cdk9 inhibition, or both. Either Cdk9 inhibition or H2Bub1 loss increased levels of antisense transcription initiating within coding regions of distinct subsets of genes; ablation of both pathways led to antisense derepression affecting over half the genome. Cdk9 and H2Bub1 cooperate to suppress antisense transcription by promoting function of the Clr6-CII histone deacetylase (HDAC) complex. H2Bub1 plays a second role, in opposition to Clr6-CII, to promote sense transcription in subtelomeric regions. Therefore, functional genomics revealed both collaborative and antagonistic functions of H2Bub1 and Cdk9.


Cancer Research | 2015

Abstract 2450: Identifying novel substrates of PLK2 using a chemical genetics approach

Poornima Ramkumar; Rebecca S. Levin; Miriam Sansó; Shashidhar S. Jatiani; Arvin C. Dar; Robert P. Fisher; Kevan M. Shokat; E. Premkumar Reddy

Polo like kinase 2 (PLK2/SNK) is a Ser/Thr kinase with roles identified in G1-S phase transition and in centriole duplication prior to entry into mitosis. PLK2 null embryonic fibroblasts have a slower proliferative rate and delayed entry into S-phase than their wild-type counterparts validating its role in cell cycle. Furthermore, wild-type p53, in response to DNA damage, induces PLK2 expression and checkpoint arrest. While these studies suggest that PLK2 is a tumor suppressor, studies have also shown that PLK2 binds to and phosphorylates mutant p53, but not wild-type p53, thereby promoting an oncogenic, auto-regulatory feedback loop, suggesting a potential role for PLK2 in proliferation. These observations suggest that PLK2 can function as an oncogene as well as a tumor suppressor. It is likely that cellular context and the nature of substrates are key to our understanding of the role that PLK2 plays in tumorigenesis. Although these reports highlight the importance of PLK2 in a variety of cellular processes, there is limited knowledge on the various proteins that are regulated by PLK2. We have optimized a chemical genetics system to identify novel substrates of PLK2. To that end, we mutated the conserved gatekeeper residue in the kinase domain of PLK2 (Leu159) to a smaller amino acid (Gly) to enlarge the ATP binding pocket. This engineered kinase, L159G-PLK2 (Plk2-as) is now able to utilize ATP analogs that carry a N6 modified bulky substituent of nucleoside residue such as N6-Phenylethyl ATP to phosphorylate substrates. We took advantage of this system to tag and isolate PLK2 substrates in vitro. Wild type and analog sensitive-PLK2 kinase domains were purified from bacteria and conditions were optimized for specific thio-phosphorylation of cell lysates by PLK2-as kinase. Labeling of cell lysates and subsequent mass spectrometry identified Signal transducer and activator of transcription 1 (STAT1) as a novel substrate of PLK2. In vitro PLK2 kinase assays in the presence or absence of specific PLK2 inhibitor, ON1231320, validated STAT1 to be a novel substrate of PLK2. Since mass spectrometry identified Ser708 as the potential phosphorylation site, we designed WT and mutant S708A peptides that span 16 amino acids of the phosphorylation site and used them as substrates for PLK1 and PLK2 in vitro kinase assays. While PLK2 readily phosphorylated the WT peptide, phosphorylation of the S708A mutant peptide was significantly reduced suggesting that PLK2 phosphorylates STAT1 at Ser708. This was further confirmed by our observation that ON1231320, a PLK2- specific inhibitor, inhibited PLK2-mediated phosphorylation of WT peptide. In addition, neither the WT nor the S708A peptide was phosphorylated by PLK1, indicating that Ser708 is a PLK2-specific phosphorylation site. Together, these studies validate that STAT1 is a bona fide target of PLK2. Citation Format: Poornima Ramkumar, Rebecca S. Levin, Miriam Sanso, Shashidhar Jatiani, Arvin C. Dar, Robert P. Fisher, Kevan M. Shokat, E Premkumar Reddy. Identifying novel substrates of PLK2 using a chemical genetics approach. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2450. doi:10.1158/1538-7445.AM2015-2450

Collaboration


Dive into the Miriam Sansó's collaboration.

Top Co-Authors

Avatar

Robert P. Fisher

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pabitra K Parua

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chao Zhang

University of California

View shared research outputs
Top Co-Authors

Avatar

Stéphane Larochelle

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bradley Benjamin

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge