131 results on '"Tom Moss"'
Search Results
2. HMG-boxes, ribosomopathies and neurodegenerative disease
- Author
-
Tom Moss, Mark S. LeDoux, and Colyn Crane-Robinson
- Subjects
neuroregression syndrome ,ribosomopathy ,upstream binding factor (UBF/UBTF) ,ribosome biogenesis ,RNA polymerase I (POLR1) ,TBP-TAF complex SL1 ,Genetics ,QH426-470 - Abstract
The UBTF E210K neuroregression syndrome is a predominantly neurological disorder caused by recurrent de novo dominant variants in Upstream Binding Factor, that is, essential for transcription of the ribosomal RNA genes. This unusual form of ribosomopathy is characterized by a slow decline in cognition, behavior, and sensorimotor functioning during the critical period of development. UBTF (or UBF) is a multi-HMGB-box protein that acts both as an epigenetic factor to establish “open” chromatin on the ribosomal genes and as a basal transcription factor in their RNA Polymerase I transcription. Here we review the possible mechanistic connections between the UBTF variants, ribosomal RNA gene transcription and the neuroregression syndrome, and suggest that DNA topology may play an important role.
- Published
- 2023
- Full Text
- View/download PDF
3. Ribosomal DNA promoter recognition is determined in vivo by cooperation between UBTF1 and SL1 and is compromised in the UBTF-E210K neuroregression syndrome.
- Author
-
Michel G Tremblay, Dany S Sibai, Melissa Valère, Jean-Clément Mars, Frédéric Lessard, Roderick T Hori, Mohammad Moshahid Khan, Victor Y Stefanovsky, Mark S LeDoux, and Tom Moss
- Subjects
Genetics ,QH426-470 - Abstract
Transcription of the ~200 mouse and human ribosomal RNA genes (rDNA) by RNA Polymerase I (RPI/PolR1) accounts for 80% of total cellular RNA, around 35% of all nuclear RNA synthesis, and determines the cytoplasmic ribosome complement. It is therefore a major factor controlling cell growth and its misfunction has been implicated in hypertrophic and developmental disorders. Activation of each rDNA repeat requires nucleosome replacement by the architectural multi-HMGbox factor UBTF to create a 15.7 kbp nucleosome free region (NFR). Formation of this NFR is also essential for recruitment of the TBP-TAFI factor SL1 and for preinitiation complex (PIC) formation at the gene and enhancer-associated promoters of the rDNA. However, these promoters show little sequence commonality and neither UBTF nor SL1 display significant DNA sequence binding specificity, making what drives PIC formation a mystery. Here we show that cooperation between SL1 and the longer UBTF1 splice variant generates the specificity required for rDNA promoter recognition in cell. We find that conditional deletion of the TAF1B subunit of SL1 causes a striking depletion of UBTF at both rDNA promoters but not elsewhere across the rDNA. We also find that while both UBTF1 and -2 variants bind throughout the rDNA NFR, only UBTF1 is present with SL1 at the promoters. The data strongly suggest an induced-fit model of RPI promoter recognition in which UBTF1 plays an architectural role. Interestingly, a recurrent UBTF-E210K mutation and the cause of a pediatric neurodegeneration syndrome provides indirect support for this model. E210K knock-in cells show enhanced levels of the UBTF1 splice variant and a concomitant increase in active rDNA copies. In contrast, they also display reduced rDNA transcription and promoter recruitment of SL1. We suggest the underlying cause of the UBTF-E210K syndrome is therefore a reduction in cooperative UBTF1-SL1 promoter recruitment that may be partially compensated by enhanced rDNA activation.
- Published
- 2022
- Full Text
- View/download PDF
4. A Deconvolution Protocol for ChIP-Seq Reveals Analogous Enhancer Structures on the Mouse and Human Ribosomal RNA Genes
- Author
-
Jean-Clement Mars, Marianne Sabourin-Felix, Michel G. Tremblay, and Tom Moss
- Subjects
ChIP-Seq deconvolution ,RNA polymerase I (RPI, PolI, Polr1) ,ribosomal RNA (rRNA) genes ,upstream binding factor (UBF/UBTF) ,selectivity factor SL1 ,Genetics ,QH426-470 - Abstract
The combination of Chromatin Immunoprecipitation and Massively Parallel Sequencing, or ChIP-Seq, has greatly advanced our genome-wide understanding of chromatin and enhancer structures. However, its resolution at any given genetic locus is limited by several factors. In applying ChIP-Seq to the study of the ribosomal RNA genes, we found that a major limitation to resolution was imposed by the underlying variability in sequence coverage that very often dominates the protein–DNA interaction profiles. Here, we describe a simple numerical deconvolution approach that, in large part, corrects for this variability, and significantly improves both the resolution and quantitation of protein–DNA interaction maps deduced from ChIP-Seq data. This approach has allowed us to determine the in vivo organization of the RNA polymerase I preinitiation complexes that form at the promoters and enhancers of the mouse (Mus musculus) and human (Homo sapiens) ribosomal RNA genes, and to reveal a phased binding of the HMG-box factor UBF across the rDNA. The data identify and map a “Spacer Promoter” and associated stalled polymerase in the intergenic spacer of the human ribosomal RNA genes, and reveal a very similar enhancer structure to that found in rodents and lower vertebrates.
- Published
- 2018
- Full Text
- View/download PDF
5. MECP2 Mutation Interrupts Nucleolin–mTOR–P70S6K Signaling in Rett Syndrome Patients
- Author
-
Carl O. Olson, Shervin Pejhan, Daniel Kroft, Kimia Sheikholeslami, David Fuss, Marjorie Buist, Annan Ali Sher, Marc R. Del Bigio, Yehezkel Sztainberg, Victoria Mok Siu, Lee Cyn Ang, Marianne Sabourin-Felix, Tom Moss, and Mojgan Rastegar
- Subjects
MECP2 mutations ,Rett syndrome ,human brain tissues ,DNA methylation ,ribosome biogenesis ,mTOR ,Genetics ,QH426-470 - Abstract
Rett syndrome (RTT) is a severe and rare neurological disorder that is caused by mutations in the X-linked MECP2 (methyl CpG-binding protein 2) gene. MeCP2 protein is an important epigenetic factor in the brain and in neurons. In Mecp2-deficient neurons, nucleoli structures are compromised. Nucleoli are sites of active ribosomal RNA (rRNA) transcription and maturation, a process mainly controlled by nucleolin and mechanistic target of rapamycin (mTOR)–P70S6K signaling. Currently, it is unclear how nucleolin–rRNA–mTOR–P70S6K signaling from RTT cellular model systems translates into human RTT brain. Here, we studied the components of nucleolin–rRNA–mTOR–P70S6K signaling in the brain of RTT patients with common T158M and R255X mutations. Immunohistochemical examination of T158M brain showed disturbed nucleolin subcellular localization, which was absent in Mecp2-deficient homozygous male or heterozygote female mice, compared to wild type (WT). We confirmed by Western blot analysis that nucleolin protein levels are altered in RTT brain, but not in Mecp2-deficient mice. Further, we studied the expression of rRNA transcripts in Mecp2-deficient mice and RTT patients, as downstream molecules that are controlled by nucleolin. By data mining of published ChIP-seq studies, we showed MeCP2-binding at the multi-copy rRNA genes in the mouse brain, suggesting that rRNA might be a direct MeCP2 target gene. Additionally, we observed compromised mTOR–P70S6K signaling in the human RTT brain, a molecular pathway that is upstream of rRNA–nucleolin molecular conduits. RTT patients showed significantly higher phosphorylation of active mTORC1 or mTORC2 complexes compared to age- and sex-matched controls. Correlational analysis of mTORC1/2–P70S6K signaling pathway identified multiple points of deviation from the control tissues that may result in abnormal ribosome biogenesis in RTT brain. To our knowledge, this is the first report of deregulated nucleolin–rRNA–mTOR–P70S6K signaling in the human RTT brain. Our results provide important insight toward understanding the molecular properties of human RTT brain.
- Published
- 2018
- Full Text
- View/download PDF
6. A unique enhancer boundary complex on the mouse ribosomal RNA genes persists after loss of Rrn3 or UBF and the inactivation of RNA polymerase I transcription.
- Author
-
Chelsea Herdman, Jean-Clement Mars, Victor Y Stefanovsky, Michel G Tremblay, Marianne Sabourin-Felix, Helen Lindsay, Mark D Robinson, and Tom Moss
- Subjects
Genetics ,QH426-470 - Abstract
Transcription of the several hundred of mouse and human Ribosomal RNA (rRNA) genes accounts for the majority of RNA synthesis in the cell nucleus and is the determinant of cytoplasmic ribosome abundance, a key factor in regulating gene expression. The rRNA genes, referred to globally as the rDNA, are clustered as direct repeats at the Nucleolar Organiser Regions, NORs, of several chromosomes, and in many cells the active repeats are transcribed at near saturation levels. The rDNA is also a hotspot of recombination and chromosome breakage, and hence understanding its control has broad importance. Despite the need for a high level of rDNA transcription, typically only a fraction of the rDNA is transcriptionally active, and some NORs are permanently silenced by CpG methylation. Various chromatin-remodelling complexes have been implicated in counteracting silencing to maintain rDNA activity. However, the chromatin structure of the active rDNA fraction is still far from clear. Here we have combined a high-resolution ChIP-Seq protocol with conditional inactivation of key basal factors to better understand what determines active rDNA chromatin. The data resolve questions concerning the interdependence of the basal transcription factors, show that preinitiation complex formation is driven by the architectural factor UBF (UBTF) independently of transcription, and that RPI termination and release corresponds with the site of TTF1 binding. They further reveal the existence of an asymmetric Enhancer Boundary Complex formed by CTCF and Cohesin and flanked upstream by phased nucleosomes and downstream by an arrested RNA Polymerase I complex. We find that the Enhancer Boundary Complex is the only site of active histone modification in the 45kbp rDNA repeat. Strikingly, it not only delimits each functional rRNA gene, but also is stably maintained after gene inactivation and the re-establishment of surrounding repressive chromatin. Our data define a poised state of rDNA chromatin and place the Enhancer Boundary Complex as the likely entry point for chromatin remodelling complexes.
- Published
- 2017
- Full Text
- View/download PDF
7. The endocytic adapter E-Syt2 recruits the p21 GTPase activated kinase PAK1 to mediate actin dynamics and FGF signalling
- Author
-
Steve Jean, Michel G. Tremblay, Chelsea Herdman, François Guillou, and Tom Moss
- Subjects
Extended synaptotagmin ,E-Syt2 ,p21-GTPase activated kinase ,PAK1 ,Cortical actin ,FGF receptor ,FGF signalling ,Endocytosis ,Science ,Biology (General) ,QH301-705.5 - Abstract
Summary Fibroblast growth factor (FGF) signalling plays an essential role in early vertebrate development. However, the response to FGF requires endocytosis of the activated FGF receptor (FGFR) that is in part dependent on remodelling of the actin cytoskeleton. Recently we showed that the extended synaptotagmin family plasma membrane protein, E-Syt2, is an essential endocytic adapter for FGFR1. Here we show E-Syt2 is also an interaction partner for the p21-GTPase Activated Kinase PAK1. The phospholipid binding C2C domain of E-Syt2 specifically binds a site adjacent to the CRIB/GBD of PAK1. PAK1 and E-Syt2 selectively complex with FGFR1 and functionally cooperate in the FGF signalling. E-Syt2 binding suppresses actin polymerization and inhibits the activation of PAK1 by the GTPases Cdc42 and Rac. Interestingly, the E-Syt2 binding site on PAK1 extensively overlaps a site recently suggested to bind phospholipids. Our data suggest that PAK1 interacts with phospholipid membrane domains via E-Syt2, where it may cooperate in the E-Syt2-dependent endocytosis of activated FGFR1 by modulating cortical actin stability.
- Published
- 2012
- Full Text
- View/download PDF
8. Conditional inactivation of Upstream Binding Factor reveals its epigenetic functions and the existence of a somatic nucleolar precursor body.
- Author
-
Nourdine Hamdane, Victor Y Stefanovsky, Michel G Tremblay, Attila Németh, Eric Paquet, Frédéric Lessard, Elaine Sanij, Ross Hannan, and Tom Moss
- Subjects
Genetics ,QH426-470 - Abstract
Upstream Binding Factor (UBF) is a unique multi-HMGB-box protein first identified as a co-factor in RNA polymerase I (RPI/PolI) transcription. However, its poor DNA sequence selectivity and its ability to generate nucleosome-like nucleoprotein complexes suggest a more generalized role in chromatin structure. We previously showed that extensive depletion of UBF reduced the number of actively transcribed ribosomal RNA (rRNA) genes, but had little effect on rRNA synthesis rates or cell proliferation, leaving open the question of its requirement for RPI transcription. Using gene deletion in mouse, we now show that UBF is essential for embryo development beyond morula. Conditional deletion in cell cultures reveals that UBF is also essential for transcription of the rRNA genes and that it defines the active chromatin conformation of both gene and enhancer sequences. Loss of UBF prevents formation of the SL1/TIF1B pre-initiation complex and recruitment of the RPI-Rrn3/TIF1A complex. It is also accompanied by recruitment of H3K9me3, canonical histone H1 and HP1α, but not by de novo DNA methylation. Further, genes retain penta-acetyl H4 and H2A.Z, suggesting that even in the absence of UBF the rRNA genes can maintain a potentially active state. In contrast to canonical histone H1, binding of H1.4 is dependent on UBF, strongly suggesting that it plays a positive role in gene activity. Unexpectedly, arrest of rRNA synthesis does not suppress transcription of the 5S, tRNA or snRNA genes, nor expression of the several hundred mRNA genes implicated in ribosome biogenesis. Thus, rRNA gene activity does not coordinate global gene expression for ribosome biogenesis. Loss of UBF also unexpectedly induced the formation in cells of a large sub-nuclear structure resembling the nucleolar precursor body (NPB) of oocytes and early embryos. These somatic NPBs contain rRNA synthesis and processing factors but do not associate with the rRNA gene loci (NORs).
- Published
- 2014
- Full Text
- View/download PDF
9. Agonistic and antagonistic roles for TNIK and MINK in non-canonical and canonical Wnt signalling.
- Author
-
Alexander Mikryukov and Tom Moss
- Subjects
Medicine ,Science - Abstract
Wnt signalling is a key regulatory factor in animal development and homeostasis and plays an important role in the establishment and progression of cancer. Wnt signals are predominantly transduced via the Frizzled family of serpentine receptors to two distinct pathways, the canonical ß-catenin pathway and a non-canonical pathway controlling planar cell polarity and convergent extension. Interference between these pathways is an important determinant of cellular and phenotypic responses, but is poorly understood. Here we show that TNIK (Traf2 and Nck-interacting kinase) and MINK (Misshapen/NIKs-related kinase) MAP4K signalling kinases are integral components of both canonical and non-canonical pathways in Xenopus. xTNIK and xMINK interact and are proteolytically cleaved in vivo to generate Kinase domain fragments that are active in signal transduction, and Citron-NIK-Homology (CNH) Domain fragments that are suppressive. The catalytic activity of the Kinase domain fragments of both xTNIK and xMINK mediate non-canonical signalling. However, while the Kinase domain fragments of xTNIK also mediate canonical signalling, the analogous fragments derived from xMINK strongly antagonize this signalling. Our data suggest that the proteolytic cleavage of xTNIK and xMINK determines their respective activities and is an important factor in controlling the balance between canonical and non-canonical Wnt signalling in vivo.
- Published
- 2012
- Full Text
- View/download PDF
10. ALegionellatoxin mimics tRNA and glycosylates the translation machinery to trigger a ribotoxic stress response
- Author
-
Advait Subramanian, Lan Wang, Tom Moss, Mark Voorhies, Smriti Sangwan, Erica Stevenson, Ernst H. Pulido, Samentha Kwok, Nevan J. Krogan, Danielle L. Swaney, Stephen N. Floor, Anita Sil, Peter Walter, and Shaeri Mukherjee
- Abstract
Pathogens often secrete proteins or nucleic acids that mimic the structure and/or function of molecules expressed in their hosts. Molecular mimicry empowers pathogens to subvert critical host processes and establish infection. We report that the intracellular bacteriumLegionella pneumophilasecretes the toxin SidI (substrate of icm/dot transporter I), which possesses a transfer RNA (tRNA)-like shape and functions as a mannosyl transferase. The 3.1 Å cryo-EM structure of SidI reveals an N-terminal domain that exhibits a characteristic ‘inverted L-shape’ and charge distribution that is present in two other known protein mimics of tRNAs, the bacterial elongation factor EF-G and the mammalian release factor eRF1. In addition, we show that SidI’s C-terminal domain adopts a glycosyl transferase B fold similar to a mannosyl transferase. This molecular coupling of the protein’s fold and enzymatic function allows SidI to bind and glycosylate components of the host translation apparatus, including the ribosome, resulting in a robust block of protein synthesis that is comparable in potency to ricin, one of the most powerful toxins known. Additionally, we find that translational pausing activated by SidI elicits a stress response signature reminiscent of the ribotoxic stress response that is activated by elongation inhibitors that induce ribosome collisions. SidI-mediated effects on the ribosome activate the stress kinases ZAKα and p38, which in turn drive an accumulation of the protein activating transcription factor 3 (ATF3). Intriguingly, ATF3 escapes the translation block imposed by SidI, translocates to the nucleus, and orchestrates the transcription of stress-inducible genes that promote cell death. Thus, usingLegionellaand its effectors as tools, we have unravelled the role of a ribosome-to-nuclear signalling pathway that regulates cell fate.
- Published
- 2022
- Full Text
- View/download PDF
11. Behavioral and molecular effects of Ubtf knockout and knockdown in mice
- Author
-
Roderick T. Hori, Mohammad Moshahid Khan, Jianfeng Xiao, Phillip W. Hargrove, Tom Moss, and Mark S. LeDoux
- Subjects
Adult ,Mice, Knockout ,General Neuroscience ,Infant ,Mice, Inbred C57BL ,Disease Models, Animal ,Mice ,Mutation ,Animals ,Humans ,Neurology (clinical) ,Nervous System Diseases ,Molecular Biology ,Developmental Biology - Abstract
The UBTF E210K neuroregression syndrome is caused by de novo dominant mutations in UBTF (NM_014233.3:c.628G A, p.Glu210Lys). In humans, onset is typically at 2.5 to 3 years and characterized by slow progression of global motor, cognitive and behavioral dysfunction. Other potentially pathogenic UBTF variants have been reported in humans with severe neurological disease and it remains undetermined if the UBTF E210K mutation operates via gain- and/or loss-of-function. Here we examine the behavioral, cognitive, motor, and molecular effects of Ubtf knockout and knockdown in mice as a means of gauging the role of loss-of-function in humans. Ubtf
- Published
- 2022
12. Control of Ribosomal RNA Synthesis by Hematopoietic Transcription Factors
- Author
-
Charles Antony, Subin S. George, Justin Blum, Patrick Somers, Chelsea L. Thorsheim, Dexter J. Wu-Corts, Yuxi Ai, Long Gao, Kaosheng Lv, Michel G. Tremblay, Tom Moss, Kai Tan, Jeremy E. Wilusz, Austen R. D. Ganley, Maxim Pimkin, and Vikram R. Paralkar
- Subjects
Mice ,Transcription, Genetic ,RNA Polymerase I ,RNA, Ribosomal ,Humans ,Animals ,RNA ,Cell Biology ,DNA, Ribosomal ,Molecular Biology ,Chromatin ,Transcription Factors - Abstract
SUMMARYRibosomal RNAs (rRNAs) are the most abundant cellular RNAs, and their synthesis from rDNA repeats by RNA Polymerase I accounts for the bulk of all transcription. Despite substantial variation in rRNA transcription rates across cell types, little is known about cell-type-specific factors that bind rDNA and regulate rRNA transcription to meet tissue-specific needs. Using hematopoiesis as a model system, we mapped about 2200 ChIP-Seq datasets for 250 transcription factors (TFs) and chromatin proteins to human and mouse rDNA, and identified robust binding of multiple TF families to canonical TF motifs on rDNA. Using a 47S-FISH-Flow assay developed for nascent rRNA quantification, we demonstrated that targeted degradation of CEBPA (C/EBP alpha), a critical hematopoietic TF with conserved rDNA binding, caused rapid reduction in rRNA transcription due to reduced Pol I occupancy. Our work identifies numerous potential rRNA regulators, and provides a template for dissection of TF roles in rRNA transcription.HIGHLIGHTSMultiple cell-type-specific transcription factors (TFs) bind canonical motifs on rDNA.The hematopoietic TF CEBPA binds to active rDNA alleles at a conserved site.CEBPA promotes Polymerase I occupancy and rRNA transcription in myeloid progenitors.We present ‘47S-FISH-Flow,’ a sensitive assay to quantify nascent rRNA.
- Published
- 2022
- Full Text
- View/download PDF
13. The human RNA polymerase I structure reveals an HMG-like transcription factor docking domain specific to metazoans
- Author
-
Julia L. Daiß, Michael Pilsl, Kristina Straub, Andrea Bleckmann, Mona Höcherl, Florian B. Heiss, Guillermo Abascal-Palacios, Ewan Ramsay, Katarina Tlučková, Jean-Clement Mars, Astrid Bruckmann, Carrie Bernecky, Valérie Lamour, Konstantin Panov, Alessandro Vannini, Tom Moss, and Christoph Engel
- Abstract
Transcription of the ribosomal RNA precursor by RNA polymerase (Pol) I is a major determinant of cellular growth and dysregulation is observed in many cancer types. Here, we present the purification of human Pol I from cells carrying a genomic GFP-fusion on the largest subunit allowing the structural and functional analysis of the enzyme across species. In contrast to yeast, human Pol I carries a single-subunit stalk and in vitro transcription indicates a reduced proofreading activity. Determination of the human Pol I cryo-EM reconstruction in a close-to-native state rationalizes the effects of disease-associated mutations and uncovers an additional domain that is built into the sequence of Pol I subunit RPA1. This ‘dock II’ domain resembles a truncated HMG-box incapable of DNA-binding which may serve as a downstream-transcription factor binding platform in metazoans. Biochemical analysis and ChIP data indicate that Topoisomerase 2a can be recruited to Pol I via the domain and cooperates with the HMG-box domain containing factor UBF. These adaptations of the metazoan Pol I transcription system may allow efficient release of positive DNA supercoils accumulating downstream of the transcription bubble.
- Published
- 2021
- Full Text
- View/download PDF
14. The chromatin landscape of the ribosomal RNA genes in mouse and human
- Author
-
Jean-Clement Mars, Tom Moss, Marianne Sabourin-Felix, and Michel G. Tremblay
- Subjects
Transcriptional Activation ,0106 biological sciences ,Transcription, Genetic ,Ribosome biogenesis ,Computational biology ,01 natural sciences ,Ribosome ,Mice ,03 medical and health sciences ,Genetics ,Animals ,Humans ,Direct repeat ,Gene Silencing ,Gene ,030304 developmental biology ,0303 health sciences ,biology ,Genes, rRNA ,Ribosomal RNA ,Chromatin ,3. Good health ,Histone ,Gene Expression Regulation ,biology.protein ,Pol1 Transcription Initiation Complex Proteins ,Chromatin immunoprecipitation ,010606 plant biology & botany - Abstract
The rRNA genes of mouse and human encode the three major RNAs of the ribosome and as such are essential for growth and development. These genes are present in high copy numbers and arranged as direct repeats at the Nucleolar Organizer Regions on multiple chromosomes. Not all the rRNA genes are transcriptionally active, but the molecular mechanisms that determine activity are complex and still poorly understood. Recent studies applying a novel Deconvolution Chromatin Immunoprecipitation (DChIP-Seq) technique in conjunction with conditional gene inactivation provide new insights into the structure of the active rRNA genes and question previous assumptions on the role of chromatin and histone modifications. We suggest an alternative model for the active rRNA gene chromatin and discuss how this structure is determined and maintained.
- Published
- 2019
- Full Text
- View/download PDF
15. Bidirectional cooperation between Ubtf1 and SL1 determines RNA Polymerase I promoter recognition in cell and is negatively affected in the UBTF-E210K neuroregression syndrome
- Author
-
Tom Moss, Jean-Clement Mars, Frédéric Lessard, Roderick T. Hori, Mohammad Moshahid Khan, Dany S. Sibai, Victor Y. Stefanovsky, Melissa Valère, Mark S. LeDoux, and Michel G. Tremblay
- Subjects
Genetics ,Transcription (biology) ,Transcription preinitiation complex ,RNA polymerase I ,RNA ,Nucleosome ,Promoter ,Biology ,Gene ,Ribosome - Abstract
Transcription of the ∼200 mouse and human ribosomal RNA genes (rDNA) by RNA Polymerase I (RPI/PolR1) accounts for 80% of total cellular RNA, around 35% of all nuclear RNA synthesis, and determines the cytoplasmic ribosome complement. It is therefore a major factor controlling cell growth and its misfunction has been implicated in hypertrophic and developmental disorders. Activation of each rDNA repeat requires nucleosome replacement by the architectural multi-HMGbox factor UBTF to create a 15kbp nucleosome free region (NFR). Formation of this NFR is also essential for recruitment of the TBP-TAFI factor SL1 and for preinitiation complex (PIC) formation at the gene and enhancer-associated promoters of the rDNA. However, these promoters show little sequence commonality and neither UBTF nor SL1 display significant DNA sequence binding specificity, making what drives PIC formation a mystery. Here we show that cooperation between SL1 and the longer UBTF1 splice variant generates the specificity required for rDNA promoter recognition in cell. We find that conditional deletion of the Taf1b subunit of SL1 causes a striking depletion UBTF at both rDNA promoters but not elsewhere across the rDNA. We also find that while both UBTF1 and −2 variants bind throughout the rDNA NFR, only UBTF1 is present with SL1 at the promoters. The data strongly suggest an induced-fit model of RPI promoter recognition in which UBTF1 plays an architectural role. Interestingly, a recurrent UBTF-E210K mutation and the cause of a pediatric neurodegeneration syndrome provides indirect support for this model. E210K knock-in cells show enhanced levels of the UBTF1 splice variant and a concomitant increase in active rDNA copies. In contrast, they also display reduced rDNA transcription and promoter recruitment of SL1. We suggest the underlying cause of the UBTF-E210K syndrome is therefore a reduction in cooperative UBTF1-SL1 promoter recruitment that may be partially compensated by enhanced rDNA activation.
- Published
- 2021
- Full Text
- View/download PDF
16. Variants of the Xenopus laevis ribosomal transcription factor xUBF are developmentally regulated by differential splicing.
- Author
-
Alain Guimond and Tom Moss
- Published
- 1992
- Full Text
- View/download PDF
17. The chemotherapeutic agent CX-5461 irreversibly blocks RNA polymerase I initiation and promoter release to cause nucleolar disruption, DNA damage and cell inviability
- Author
-
Dany S Sibai, Frédéric Lessard, Mélissa Valere, Jean-Clement Mars, Marianne Sabourin-Felix, Tom Moss, and Michel G. Tremblay
- Subjects
0301 basic medicine ,AcademicSubjects/SCI01140 ,AcademicSubjects/SCI01060 ,Chemistry ,DNA damage ,Cell ,AcademicSubjects/SCI00030 ,DNA replication ,Ribosome biogenesis ,Standard Article ,Ribosomal RNA ,AcademicSubjects/SCI01180 ,3. Good health ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Transcription (biology) ,030220 oncology & carcinogenesis ,Transcription preinitiation complex ,medicine ,RNA polymerase I ,AcademicSubjects/SCI00980 - Abstract
In the search for drugs to effectively treat cancer, the last 10 years have seen a resurgence of interest in targeting ribosome biogenesis. CX-5461 is a potential inhibitor of ribosomal RNA synthesis that is now showing promise in phase I trials as a chemotherapeutic agent for a range of malignancies. Here, we show that CX-5461 irreversibly inhibits ribosomal RNA transcription by arresting RNA polymerase I (RPI/Pol1/PolR1) in a transcription initiation complex. CX-5461 does not achieve this by preventing formation of the pre-initiation complex nor does it affect the promoter recruitment of the SL1 TBP complex or the HMGB-box upstream binding factor (UBF/UBTF). CX-5461 also does not prevent the subsequent recruitment of the initiation-competent RPI–Rrn3 complex. Rather, CX-5461 blocks promoter release of RPI–Rrn3, which remains irreversibly locked in the pre-initiation complex even after extensive drug removal. Unexpectedly, this results in an unproductive mode of RPI recruitment that correlates with the onset of nucleolar stress, inhibition of DNA replication, genome-wide DNA damage and cellular senescence. Our data demonstrate that the cytotoxicity of CX-5461 is at least in part the result of an irreversible inhibition of RPI transcription initiation and hence are of direct relevance to the design of improved strategies of chemotherapy.
- Published
- 2020
18. Dynamic proteomics profiling ofLegionella pneumophilainfection unveils modulation of the host mitochondrial stress response pathway
- Author
-
Julia Noack, Danielle L. Swaney, Tom Moss, Erica Stevenson, Shaeri Mukherjee, David Jimenez-Morales, Gwendolyn M. Jang, and Nevan J. Krogan
- Subjects
biology ,Effector ,Mitochondrial unfolded protein response ,Proteome ,Secretion ,Human pathogen ,Mitochondrion ,biology.organism_classification ,Proteomics ,Legionella pneumophila ,Cell biology - Abstract
SUMMARYThe human pathogenLegionella pneumophila (L.p.)secretes ~330 bacterial effector proteins into the host cell which interfere with numerous cellular pathways and often regulate host cell proteins through post-translational modifications. However, the cellular targets and functions of mostL.p.effectors are not known. In order to obtain a global overview of potential targets of these effectors, we analyzed the host cell proteome, ubiquitinome, and phosphoproteome duringL.p.infection. Our analysis reveals dramatic spatiotemporal changes in the host cell proteome that are dependent on the secretion of bacterial effectors. Strikingly, we show thatL.p.substantially reshapes the mitochondrial proteome and modulates mitochondrial stress response pathways such as the mitochondrial unfolded protein response (UPRmt). To our knowledge, this is the first evidence of manipulation of the UPRmtby a bacterial pathogen in mammalian cells. In addition, we have identified a previously uncharacterizedL.p.effector that is targeted to host cell mitochondria and protects mitochondrial network integrity during mitochondrial stress.
- Published
- 2020
- Full Text
- View/download PDF
19. Dynamic Proteomics Profiling of Legionella pneumophila Infection Unveils Modulation of the Host Mitochondrial Stress Response Pathway
- Author
-
Nevan J. Krogan, Danielle L. Swaney, Shaeri Mukherjee, Erica Stevenson, Tom Moss, Julia Noack, David Jimenez-Morales, and Gwendolyn M. Jang
- Subjects
Proteostasis ,Effector ,Host–pathogen interaction ,Mitochondrial unfolded protein response ,Proteome ,Integrated stress response ,Mitochondrion ,Biology ,Proteomics ,Cell biology - Abstract
The human pathogen Legionella pneumophila (L.p.) secretes ~330 bacterial effector proteins into the host cell which interfere with numerous cellular pathways and often regulate host cell proteins through post-translational modifications. However, the cellular targets and functions of most L.p. effectors are not known. In order to obtain a global overview of potential targets of these effectors, we analyzed the host cell proteome, ubiquitinome, and phosphoproteome during L.p. infection. Our analysis reveals dramatic spatiotemporal changes in the host cell proteome that are dependent on the secretion of bacterial effectors. Strikingly, we show that L.p. substantially reshapes the mitochondrial proteome and modulates mitochondrial stress response pathways such as the mitochondrial unfolded protein response (UPRmt). To our knowledge, this is the first evidence of manipulation of the UPRmt by a bacterial pathogen in mammalian cells. In addition, we have identified a previously uncharacterized L.p. effector that is targeted to host cell mitochondria and protects mitochondrial network integrity during mitochondrial stress.
- Published
- 2020
- Full Text
- View/download PDF
20. A recurrent de novo missense mutation in UBTF causes developmental neuroregression
- Author
-
May Christine V. Malicdan, Jun Tian, Lawrence T. Reiter, Chris Balak, Kevin A. Hope, Michel G. Tremblay, David R. Adams, Harper B Fauni, Jianfeng Xiao, Camilo Toro, Alexis L Franks, Roderick T. Hori, Amy Goldstein, William A. Gahl, Mark S. LeDoux, Lynne A. Wolfe, Mohammad Moshahid Khan, Tom Moss, and Cynthia J. Tifft
- Subjects
Gait Ataxia ,Male ,0301 basic medicine ,Mutation, Missense ,RNA polymerase II ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Transcription (biology) ,RNA, Ribosomal, 18S ,Genetics ,medicine ,Humans ,Missense mutation ,Molecular Biology ,Exome ,Transcription factor ,Genetics (clinical) ,030304 developmental biology ,0303 health sciences ,biology ,Dysarthria ,Articles ,General Medicine ,Corrigenda ,Magnetic Resonance Imaging ,RRNA transcription ,Hypotonia ,Pedigree ,030104 developmental biology ,Child, Preschool ,Cancer research ,biology.protein ,Muscle Hypotonia ,Female ,medicine.symptom ,Pol1 Transcription Initiation Complex Proteins ,030217 neurology & neurosurgery - Abstract
UBTF (upstream binding transcription factor) exists as two isoforms; UBTF1 regulates rRNA transcription by RNA polymerase 1, whereas UBTF2 regulates mRNA transcription by RNA polymerase 2. Herein, we describe 4 patients with very similar patterns of neuroregression due to recurrent de novo mutations in UBTF (GRCh37/hg19, NC_000017.10: g.42290219C > T, NM_014233.3: c.628G > A) resulting in the same amino acid change in both UBTF1 and UBTF2 (p.Glu210Lys [p.E210K]). Disease onset in our cohort was at 2.5 to 3 years and characterized by slow progression of global motor, cognitive and behavioral dysfunction. Notable early features included hypotonia with a floppy gait, high-pitched dysarthria and hyperactivity. Later features included aphasia, dystonia, and spasticity. Speech and ambulatory ability were lost by the early teens. Magnetic resonance imaging showed progressive generalized cerebral atrophy (supratentorial > infratentorial) with involvement of both gray and white matter. Patient fibroblasts showed normal levels of UBTF transcripts, increased expression of pre-rRNA and 18S rRNA, nucleolar abnormalities, markedly increased numbers of DNA breaks, defective cell-cycle progression, and apoptosis. Expression of mutant human UBTF1 in Drosophila neurons was lethal. Although no loss-of-function variants are reported in the Exome Aggregation Consortium (ExAC) database and Ubtf−/− is early embryonic lethal in mice, Ubtf+/− mice displayed only mild motor and behavioral dysfunction in adulthood. Our data underscore the importance of including UBTF E210K in the differential diagnosis of neuroregression and suggest that mainly gain-of-function mechanisms contribute to the pathogenesis of the UBTF E210K neuroregression syndrome.
- Published
- 2018
- Full Text
- View/download PDF
21. A Deconvolution Protocol for ChIP-Seq Reveals Analogous Enhancer Structures on the Mouse and Human Ribosomal RNA Genes
- Author
-
Tom Moss, Marianne Sabourin-Felix, Jean-Clement Mars, and Michel G. Tremblay
- Subjects
0301 basic medicine ,Chromatin Immunoprecipitation ,ChIP-Seq deconvolution ,5.8S ribosomal RNA ,Enhancer RNAs ,QH426-470 ,Investigations ,Biology ,DNA, Ribosomal ,ribosomal RNA (rRNA) genes ,Mice ,03 medical and health sciences ,RNA Polymerase I ,Sequence Homology, Nucleic Acid ,28S ribosomal RNA ,Genetics ,RNA polymerase I ,Animals ,Humans ,Promoter Regions, Genetic ,selectivity factor SL1 ,Enhancer ,Molecular Biology ,Genetics (clinical) ,Genome ,Base Sequence ,Intron ,High-Throughput Nucleotide Sequencing ,RNA ,Genes, rRNA ,Ribosomal RNA ,Enhancer Elements, Genetic ,030104 developmental biology ,RNA, Ribosomal ,RNA polymerase I (RPI, PolI, Polr1) ,upstream binding factor (UBF/UBTF) ,Pol1 Transcription Initiation Complex Proteins ,Ribosomes ,Sequence Alignment - Abstract
The combination of Chromatin Immunoprecipitation and Massively Parallel Sequencing, or ChIP-Seq, has greatly advanced our genome-wide understanding of chromatin and enhancer structures. However, its resolution at any given genetic locus is limited by several factors. In applying ChIP-Seq to the study of the ribosomal RNA genes, we found that a major limitation to resolution was imposed by the underlying variability in sequence coverage that very often dominates the protein–DNA interaction profiles. Here, we describe a simple numerical deconvolution approach that, in large part, corrects for this variability, and significantly improves both the resolution and quantitation of protein–DNA interaction maps deduced from ChIP-Seq data. This approach has allowed us to determine the in vivo organization of the RNA polymerase I preinitiation complexes that form at the promoters and enhancers of the mouse (Mus musculus) and human (Homo sapiens) ribosomal RNA genes, and to reveal a phased binding of the HMG-box factor UBF across the rDNA. The data identify and map a “Spacer Promoter” and associated stalled polymerase in the intergenic spacer of the human ribosomal RNA genes, and reveal a very similar enhancer structure to that found in rodents and lower vertebrates.
- Published
- 2018
- Full Text
- View/download PDF
22. Point mutation analysis of the Xenopus laevis RNA polymerase I core promoter.
- Author
-
S. Firek, C. Read, D. R. Smith, and Tom Moss
- Published
- 1990
- Full Text
- View/download PDF
23. Loss of all 3 Extended Synaptotagmins does not affect normal mouse development, viability or fertility
- Author
-
Michel G. Tremblay and Tom Moss
- Subjects
Male ,0301 basic medicine ,ESyt2 ,ESyt3 ,Genotype ,Embryonic Development ,Biology ,Endocytosis ,Bone and Bones ,Synaptotagmins ,Mice ,03 medical and health sciences ,Cell surface receptor ,Animals ,Lung ,Molecular Biology ,Crosses, Genetic ,C2 domain ,Developmental Expression Profiling ,E-Syt3 ,E-Syt2 ,E-Syt1 ,Extended-Synaptotagmin ,ORAI1 ,Endoplasmic reticulum ,Gene Expression Regulation, Developmental ,Membrane Proteins ,STIM1 ,Cell Biology ,Expression Profiling ,Embryo, Mammalian ,Cell biology ,Viability' Mouse ,Mutagenesis, Insertional ,Fertility ,030104 developmental biology ,Membrane protein ,Gene Targeting ,Female ,ESyt1 ,Gene Deletion ,Spleen ,Reports ,Developmental Biology - Abstract
The extended synaptotagmins, E-Syt1, 2 and 3, are multiple C2 domain membrane proteins that are tethered to the endoplasmic reticulum and interact in a calcium dependent manner with plasma membrane phospholipids to form endoplasmic reticulum - plasma membrane junctions. These junctions have been implicated in the exchange of phospholipids between the 2 organelles. The E-Syts have further been implicated in receptor signaling and endocytosis and can interact directly with fibroblast growth factor and other cell surface receptors. Despite these multiple functions, the search for a requirement in vivo has been elusive. Most recently, we found that the genes for E-Syt2 and 3 could be inactivated without effect on mouse development, viability, fertility or morphology. We have now created insertion and deletion mutations in the last of the mouse E-Syt genes. We show that E-Syt1 is specifically expressed throughout the embryonic skeleton during the early stages of chrondrogenesis in a pattern quite distinct from that of E-Syt2 or 3. Despite this, E-Syt1 is also not required for mouse development and propagation. We further show that even the combined inactivation of all 3 E-Syt genes has no effect on mouse viability or fertility in the laboratory. However, this inactivation induces an enhancement in the expression of the genes encoding Orp5/8, Orai1, STIM1 and TMEM110, endoplasmic reticulum - plasma membrane junction proteins that potentially could compensate for E-Syt loss. Given the multiple functions suggested for the E-Syts and their evolutionary conservation, our unexpected findings suggest that they may only provide a survival advantage under specific conditions that have as yet to be identified.
- Published
- 2016
- Full Text
- View/download PDF
24. MECP2 Mutation Interrupts Nucleolin–mTOR–P70S6K Signaling in Rett Syndrome Patients
- Author
-
Annan Ali Sher, Marjorie Buist, Marc R. Del Bigio, Victoria Mok Siu, Shervin Pejhan, Marianne Sabourin-Felix, Mojgan Rastegar, Tom Moss, Carl O. Olson, David Fuss, Lee Cyn Ang, Daniel Kroft, Kimia Sheikholeslami, and Yehezkel Sztainberg
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,lcsh:QH426-470 ,Ribosome biogenesis ,ribosome biogenesis ,Rett syndrome ,mTORC1 ,mTORC2 ,MECP2 ,03 medical and health sciences ,nucleolin ,medicine ,Genetics ,Mechanistic target of rapamycin ,protein translation ,PI3K/AKT/mTOR pathway ,Genetics (clinical) ,Original Research ,DNA methylation ,biology ,human brain tissues ,medicine.disease ,3. Good health ,Cell biology ,lcsh:Genetics ,030104 developmental biology ,biology.protein ,mTOR ,Molecular Medicine ,MECP2 mutations ,Nucleolin - Abstract
Rett syndrome (RTT) is a severe and rare neurological disorder that is caused by mutations in the X-linked MECP2 (methyl CpG-binding protein 2) gene. MeCP2 protein is an important epigenetic factor in the brain and in neurons. In Mecp2-deficient neurons, nucleoli structures are compromised. Nucleoli are sites of active ribosomal RNA (rRNA) transcription and maturation, a process mainly controlled by nucleolin and mechanistic target of rapamycin (mTOR)–P70S6K signaling. Currently, it is unclear how nucleolin–rRNA–mTOR–P70S6K signaling from RTT cellular model systems translates into human RTT brain. Here, we studied the components of nucleolin–rRNA–mTOR–P70S6K signaling in the brain of RTT patients with common T158M and R255X mutations. Immunohistochemical examination of T158M brain showed disturbed nucleolin subcellular localization, which was absent in Mecp2-deficient homozygous male or heterozygote female mice, compared to wild type (WT). We confirmed by Western blot analysis that nucleolin protein levels are altered in RTT brain, but not in Mecp2-deficient mice. Further, we studied the expression of rRNA transcripts in Mecp2-deficient mice and RTT patients, as downstream molecules that are controlled by nucleolin. By data mining of published ChIP-seq studies, we showed MeCP2-binding at the multi-copy rRNA genes in the mouse brain, suggesting that rRNA might be a direct MeCP2 target gene. Additionally, we observed compromised mTOR–P70S6K signaling in the human RTT brain, a molecular pathway that is upstream of rRNA–nucleolin molecular conduits. RTT patients showed significantly higher phosphorylation of active mTORC1 or mTORC2 complexes compared to age- and sex-matched controls. Correlational analysis of mTORC1/2–P70S6K signaling pathway identified multiple points of deviation from the control tissues that may result in abnormal ribosome biogenesis in RTT brain. To our knowledge, this is the first report of deregulated nucleolin–rRNA–mTOR–P70S6K signaling in the human RTT brain. Our results provide important insight toward understanding the molecular properties of human RTT brain.
- Published
- 2018
- Full Text
- View/download PDF
25. Depletion of the cisplatin targeted HMGB-box factor UBF selectively induces p53-independent apoptotic death in transformed cells
- Author
-
Tom Moss, Michel G. Tremblay, Nourdine Hamdane, Chelsea Herdman, Victor Y. Stefanovsky, and Jean-Clement Mars
- Subjects
DNA Replication ,Male ,Ribosome biogenesis ,ribosome biogenesis ,cisplatin ,Mitosis ,oncogenic stress ,Apoptosis ,Mice, Transgenic ,Cell Separation ,Biology ,Ribosome ,Mice ,Transcription (biology) ,RNA Polymerase I ,Neoplasms ,medicine ,RNA polymerase I ,Gene silencing ,Animals ,Gene Silencing ,Upstream binding factor (UBF/UBTF) ,Cell Line, Transformed ,Cell Proliferation ,Cisplatin ,Cell Death ,Cell Cycle ,Homozygote ,Cell cycle ,Ribosomal RNA ,Flow Cytometry ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,Oncology ,RNA, Ribosomal ,Cancer research ,Female ,Tumor Suppressor Protein p53 ,Pol1 Transcription Initiation Complex Proteins ,Ribosomes ,Gene Deletion ,medicine.drug ,Research Paper - Abstract
// Nourdine Hamdane 1, 2, 3 , Chelsea Herdman 1, 2 , Jean-Clement Mars 1, 2 , Victor Stefanovsky 1 , Michel G. Tremblay 1 , Tom Moss 1, 2 1 Laboratory of Growth and Development, St-Patrick Research Group in Basic Oncology, Cancer Division of the Quebec University Hospital Research Centre, Quebec, QC, Canada 2 Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Laval University, Quebec, QC, Canada 3 Present address: Inserm, U1110, Institute of Viral and Liver Diseases, Strasbourg, France Correspondence to: Tom Moss, e-mail: Tom.Moss@crhdq.ulaval.ca Keywords: Upstream binding factor (UBF/UBTF), ribosome biogenesis, oncogenic stress, apoptosis, cisplatin Received: January 23, 2015 Accepted: July 27, 2015 Published: August 07, 2015 ABSTRACT Cisplatin-DNA adducts act as strong decoys for the Upstream Binding Factor UBF (UBTF) and have been shown to inhibit transcription of the ribosomal RNA genes by RNA polymerase I. However, it is unclear if this plays a significant role in the chemotherapeutic activity of cis- or carboplatin. We find that cisplatin in fact induces a very rapid displacement of UBF from the ribosomal RNA genes and strong inhibition of ribosomal RNA synthesis, consistent with this being an important factor in its cytotoxicity. Using conditional gene deletion, we recently showed that UBF is an essential factor for transcription of the ribosomal RNA genes and for ribosome biogenesis. We now show that loss of UBF arrests cell proliferation and induces fully penetrant, rapid and synchronous apoptosis, as well as nuclear disruption and cell death, specifically in cells subjected to oncogenic stress. Apoptosis is not affected by homozygous deletion of the p53 gene and occurs equally in cells transformed by SV40 T antigens, by Myc or by a combination of Ras & Myc oncogenes. The data strongly argue that inhibition of UBF function is a major factor in the cytotoxicity of cisplatin. Hence, drug targeting of UBF may be a preferable approach to the use of the highly toxic platins in cancer therapy.
- Published
- 2015
26. A HYBRID GENRE SUPPORTS HYBRID ROLES IN COMMUNITY-UNIVERSITY COLLABORATION
- Author
-
Timothy Henningsen, Diane Chin, Ann Feldman, Caroline Gottschalk-Druschke, Tom Moss, Nadya Pittendrigh, and Stephanie Turner Reich
- Published
- 2017
- Full Text
- View/download PDF
27. A unique enhancer boundary complex on the mouse ribosomal RNA genes persists after loss of Rrn3 or UBF and the inactivation of RNA polymerase I transcription
- Author
-
Victor Y. Stefanovsky, Marianne Sabourin-Felix, Jean-Clement Mars, Helen Lindsay, Tom Moss, Chelsea Herdman, Mark D. Robinson, Michel G. Tremblay, University of Zurich, and Moss, Tom
- Subjects
0301 basic medicine ,Cancer Research ,Embryology ,Transcription, Genetic ,Gene Expression ,Enhancer RNAs ,RNA polymerase II ,Biochemistry ,Histones ,Mice ,Pregnancy ,RNA Polymerase I ,1306 Cancer Research ,Genetics (clinical) ,Cells, Cultured ,Genetics ,Mice, Knockout ,General transcription factor ,biology ,Chromosome Biology ,Chromatin Modification ,Nuclear Proteins ,Histone Modification ,10124 Institute of Molecular Life Sciences ,Chromatin ,Nucleosomes ,Nucleic acids ,Enhancer Elements, Genetic ,Ribosomal RNA ,Epigenetics ,Female ,Pol1 Transcription Initiation Complex Proteins ,Research Article ,2716 Genetics (clinical) ,Cell biology ,Cellular structures and organelles ,lcsh:QH426-470 ,DNA transcription ,03 medical and health sciences ,1311 Genetics ,DNA-binding proteins ,1312 Molecular Biology ,Nucleolus Organizer Region ,Animals ,Gene Silencing ,Enhancer ,Non-coding RNA ,Molecular Biology ,Ribosomal DNA ,Ecology, Evolution, Behavior and Systematics ,Biology and life sciences ,Pioneer factor ,Embryos ,Proteins ,Genes, rRNA ,Sequence Analysis, DNA ,Chromatin Assembly and Disassembly ,lcsh:Genetics ,1105 Ecology, Evolution, Behavior and Systematics ,030104 developmental biology ,Genetic Loci ,Transcription preinitiation complex ,biology.protein ,570 Life sciences ,RNA ,Ribosomes ,Gene Deletion ,Developmental Biology ,Transcription Factors - Abstract
Transcription of the several hundred of mouse and human Ribosomal RNA (rRNA) genes accounts for the majority of RNA synthesis in the cell nucleus and is the determinant of cytoplasmic ribosome abundance, a key factor in regulating gene expression. The rRNA genes, referred to globally as the rDNA, are clustered as direct repeats at the Nucleolar Organiser Regions, NORs, of several chromosomes, and in many cells the active repeats are transcribed at near saturation levels. The rDNA is also a hotspot of recombination and chromosome breakage, and hence understanding its control has broad importance. Despite the need for a high level of rDNA transcription, typically only a fraction of the rDNA is transcriptionally active, and some NORs are permanently silenced by CpG methylation. Various chromatin-remodelling complexes have been implicated in counteracting silencing to maintain rDNA activity. However, the chromatin structure of the active rDNA fraction is still far from clear. Here we have combined a high-resolution ChIP-Seq protocol with conditional inactivation of key basal factors to better understand what determines active rDNA chromatin. The data resolve questions concerning the interdependence of the basal transcription factors, show that preinitiation complex formation is driven by the architectural factor UBF (UBTF) independently of transcription, and that RPI termination and release corresponds with the site of TTF1 binding. They further reveal the existence of an asymmetric Enhancer Boundary Complex formed by CTCF and Cohesin and flanked upstream by phased nucleosomes and downstream by an arrested RNA Polymerase I complex. We find that the Enhancer Boundary Complex is the only site of active histone modification in the 45kbp rDNA repeat. Strikingly, it not only delimits each functional rRNA gene, but also is stably maintained after gene inactivation and the re-establishment of surrounding repressive chromatin. Our data define a poised state of rDNA chromatin and place the Enhancer Boundary Complex as the likely entry point for chromatin remodelling complexes.
- Published
- 2017
28. A novel role for the Pol I transcription factor UBTF in maintaining genome stability through the regulation of highly transcribed Pol II genes
- Author
-
Izhak Haviv, Elaine Sanij, Jason Ellul, Amardeep S. Dhillon, Gretchen Poortinga, Tom Moss, Lawrence I. Rothblum, Analia Lesmana, Jeannine Diesch, Nourdine Hamdane, Ross D. Hannan, Richard B. Pearson, Gregory J. Goodall, Nadine Hein, Grace E. Lidgerwood, Lee H. Wong, Donald P. Cameron, Sanjie, Elaine, Diesch, Jeannine, Lesmana, Analia, Poortinga, Gretchen, Hein, Nadine, Lidgerwood, Grace, Cameron, Donald P, Ellul, Jason, Goodall, Gregory J, Wong, Lee H, Dhillon, Amardeep S, Hamdane, Nourdine, Rothblum, Lawrence I, Pearson, Richard B, Haviv, Izhak, Moss, Tom, and Hannan, Ross D
- Subjects
Chromatin Immunoprecipitation ,Transcription, Genetic ,RNA polymerase II ,Genomic Instability ,Histones ,Mice ,03 medical and health sciences ,0302 clinical medicine ,UBF ,RNA Polymerase I ,Genetics ,Transcriptional regulation ,Animals ,Humans ,Nucleosome ,Transcription factor ,Genetics (clinical) ,Cell Line, Transformed ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Binding Sites ,cellular homeostasis and growth ,biology ,Research ,Computational Biology ,High-Throughput Nucleotide Sequencing ,Promoter ,Chromatin ,Nucleosomes ,Histone ,Gene Expression Regulation ,RNA polymerase ,Gene Knockdown Techniques ,Multigene Family ,030220 oncology & carcinogenesis ,NIH 3T3 Cells ,biology.protein ,RNA Polymerase II ,Transcription Initiation Site ,Pol1 Transcription Initiation Complex Proteins ,Chromatin immunoprecipitation ,DNA Damage ,Protein Binding - Abstract
Mechanisms to coordinate programs of highly transcribed genes required for cellular homeostasis and growth are unclear. Upstream binding transcription factor (UBTF, also called UBF) is thought to function exclusively in RNA polymerase I (Pol I)-specific transcription of the ribosomal genes. Here, we report that the two isoforms of UBTF (UBTF1/2) are also enriched at highly expressed Pol II-transcribed genes throughout the mouse genome. Further analysis of UBTF1/2 DNA binding in immortalized human epithelial cells and their isogenically matched transformed counterparts reveals an additional repertoire of UBTF1/2-bound genes involved in the regulation of cell cycle checkpoints and DNA damage response. As proof of a functional role for UBTF1/2 in regulating Pol II transcription, we demonstrate that UBTF1/2 is required for recruiting Pol II to the highly transcribed histone gene clusters and for their optimal expression. Intriguingly, lack of UBTF1/2 does not affect chromatin marks or nucleosome density at histone genes. Instead, it results in increased accessibility of the histone promoters and transcribed regions to micrococcal nuclease, implicating UBTF1/2 in mediating DNA accessibility. Unexpectedly, UBTF2, which does not function in Pol I transcription, is sufficient to regulate histone gene expression in the absence of UBTF1. Moreover, depletion of UBTF1/2 and subsequent reduction in histone gene expression is associated with DNA damage and genomic instability independent of Pol I transcription. Thus, we have uncovered a novel role for UBTF1 and UBTF2 in maintaining genome stability through coordinating the expression of highly transcribed Pol I (UBTF1 activity) and Pol II genes (UBTF2 activity). Refereed/Peer-reviewed
- Published
- 2014
- Full Text
- View/download PDF
29. Loss of Extended Synaptotagmins ESyt2 and ESyt3 does not affect mouse development or viability, but in vitro cell migration and survival under stress are affected
- Author
-
Tom Moss, Michel G. Tremblay, Chelsea Herdman, and Prakash K. Mishra
- Subjects
Cell Survival ,Longevity ,Xenopus ,Embryonic Development ,Biology ,Synaptotagmins ,Mice ,Cell Cycle News & Views ,Cell Movement ,Stress, Physiological ,Animals ,Fetal Viability ,Cell adhesion ,Molecular Biology ,Embryonic Stem Cells ,Mice, Knockout ,Endoplasmic reticulum ,Calcium-Binding Proteins ,Wild type ,Membrane Proteins ,Cell migration ,Cell Biology ,Fibroblasts ,biology.organism_classification ,Molecular biology ,Cell biology ,Membrane protein ,Signal transduction ,Gene Deletion ,Developmental Biology - Abstract
The Extended Synaptotagmins (Esyts) are a family of multi-C2 domain membrane proteins with orthologs in organisms from yeast to human. Three Esyt genes exist in mouse and human and these have most recently been implicated in the formation of junctions between endoplasmic reticulum and plasma membrane, as well as the Ca(2+) dependent replenishment of membrane phospholipids. The data are consistent with a function in extracellular signal transduction and cell adhesion, and indeed Esyt2 was previously implicated in both these functions in Xenopus. Despite this, little is known of the function of the Esyts in vivo. We have generated mouse lines carrying homozygous deletions in one or both of the genes encoding the highly homologous Esyt2 and Esyt3 proteins. Surprisingly, esyt2(-/-)/esyt3(-/-) mice develop normally and are both viable and fertile. In contrast, esyt2(-/-)/esyt3(-/-) mouse embryonic fibroblasts display a reduced ability to migrate in standard in vitro assays, and are less resistant to stringent culture conditions and to oxidative stress than equivalent wild type fibroblasts.
- Published
- 2014
- Full Text
- View/download PDF
30. Metabolic Labeling in the Study of Mammalian Ribosomal RNA Synthesis
- Author
-
Victor Y, Stefanovsky and Tom, Moss
- Subjects
Electrophoresis, Agar Gel ,Mammals ,Mice ,Transcription Elongation, Genetic ,Staining and Labeling ,Transcription, Genetic ,RNA Polymerase I ,RNA, Ribosomal ,NIH 3T3 Cells ,RNA Precursors ,Animals - Abstract
RNA metabolic labeling is a method of choice in the study of dynamic changes in the rate of gene transcription and RNA processing. It is particularly applicable to transcription of the ribosomal RNA genes and their processing products due to the very high levels of ribosomal RNA synthesis. Metabolic labeling can detect changes in ribosomal RNA transcription that occur within a few minutes as opposed to the still widely used RT-PCR or Northern blot procedures that measure RNA pool sizes and at best are able to detect changes occurring over several hours or several days. Here, we describe a metabolic labeling technique applicable to the measurement of ribosomal RNA synthesis and processing rates, as well as to the determination of RNA Polymerase I transcription elongation rates.
- Published
- 2016
31. Disruption of the UBF gene induces aberrant somatic nucleolar bodies and disrupts embryo nucleolar precursor bodies
- Author
-
Victor Y. Stefanovsky, Nourdine Hamdane, Attila Németh, Tom Moss, Michel G. Tremblay, and Stefan Dillinger
- Subjects
0301 basic medicine ,030102 biochemistry & molecular biology ,Nucleolus ,Heterochromatin ,Somatic cell ,Ribosome biogenesis ,General Medicine ,Ribosomal RNA ,Biology ,Embryo, Mammalian ,Molecular biology ,Chromosome segregation ,03 medical and health sciences ,Mice ,030104 developmental biology ,Genetics ,Animals ,Gene ,Reprogramming ,Pol1 Transcription Initiation Complex Proteins ,Cell Nucleolus ,In Situ Hybridization, Fluorescence ,Cell Line, Transformed - Abstract
The nucleolus is the site of ribosome biogenesis and forms around the actively transcribed ribosomal RNA (rRNA) genes. However, the nucleolus is also implicated in cell cycle regulation, tumour suppression and chromosome segregation and nucleolar disfunction is linked to a wide range of human diseases. Interestingly, the nucleolus is also required for genome reprogramming and the establishment of heterochromatin in the mammalian embryo. Mammalian oocytes contain a subnuclear structure that is believed to be the precursor of the functional nucleolus, the Nucleolar Precursor Body (NPB). But the NPB is also required for the organisation of the zygotic heterochromatin and the establishment of pluripotency. We found that disruption of the mouse Upstream Binding Factor (UBF (UBTF)) gene caused disassembly of somatic nucleoli and the accumulation of the key rRNA gene transcription factors into dense subnuclear foci resembling NPBs. Here we show that UBF deletion causes the rRNA genes to collapse onto their centromere-proximal chromosomal sites spatially distinct from NPB-like structures, and that these structures contain rRNA gene transcription factors but not all nucleolar proteins. We further find that embryonic NPBs and their surrounding heterochromatin are both disrupted in UBF-null mouse embryos. These embryos also display subnuclear foci containing the rRNA gene transcription factors and arrest development before completing the forth cleavage division. The data suggest that the rRNA gene transcription factors have an intrinsic ability to interact and form a discrete nuclear compartment even in the absence of any rRNA gene activity and that the formation or maintenance of the zygotic NPB and surrounding heterochromatin requires UBF.
- Published
- 2016
32. Extended-Synaptotagmins (E-Syts); the extended story
- Author
-
Tom Moss and Chelsea Herdman
- Subjects
0301 basic medicine ,Pharmacology ,Protein Conformation ,Computational biology ,Biology ,Bioinformatics ,Conserved sequence ,Synaptotagmins ,03 medical and health sciences ,030104 developmental biology ,Protein structure ,Membrane protein ,Animals ,Humans ,Signal transduction ,Lipid Transport ,Organism ,Calcium signaling ,Signal Transduction - Abstract
The Extended-Synaptotagmin (E-Syt) membrane proteins were only recently discovered, but have already been implicated in a range of interrelated cellular functions, including calcium and receptor signaling, and membrane lipid transport. However, despite their evolutionary conservation and detailed studies of their molecular actions, we still have little idea of how and when these proteins are required in cellular and organism physiology. Here we review our present understanding of the E-Syts and discuss the molecular functions and in vivo requirements for these proteins.
- Published
- 2016
33. Metabolic Labeling in the Study of Mammalian Ribosomal RNA Synthesis
- Author
-
Tom Moss and Victor Y. Stefanovsky
- Subjects
0301 basic medicine ,5.8S ribosomal RNA ,RNA ,Biology ,Ribosomal RNA ,Molecular biology ,18S ribosomal RNA ,03 medical and health sciences ,030104 developmental biology ,Biochemistry ,Transcription (biology) ,28S ribosomal RNA ,RNA polymerase I ,30S - Abstract
RNA metabolic labeling is a method of choice in the study of dynamic changes in the rate of gene transcription and RNA processing. It is particularly applicable to transcription of the ribosomal RNA genes and their processing products due to the very high levels of ribosomal RNA synthesis. Metabolic labeling can detect changes in ribosomal RNA transcription that occur within a few minutes as opposed to the still widely used RT-PCR or Northern blot procedures that measure RNA pool sizes and at best are able to detect changes occurring over several hours or several days. Here, we describe a metabolic labeling technique applicable to the measurement of ribosomal RNA synthesis and processing rates, as well as to the determination of RNA Polymerase I transcription elongation rates.
- Published
- 2016
- Full Text
- View/download PDF
34. The p21-activated kinase Pak1 regulates induction and migration of the neural crest in Xenopus
- Author
-
Tom Moss, Doris Wedlich, and Nicolas Bisson
- Subjects
Cellular differentiation ,Xenopus ,Xenopus laevis ,Cranial neural crest ,PAK1 ,Cell Movement ,Animals ,Molecular Biology ,Embryonic Induction ,biology ,Twist-Related Protein 1 ,Neural crest ,Cell Differentiation ,SOX9 Transcription Factor ,Cell migration ,Cell Biology ,biology.organism_classification ,Cell biology ,Transplantation ,p21-Activated Kinases ,Neural Crest ,Mutation ,Cancer research ,Snail Family Transcription Factors ,Signal transduction ,Signal Transduction ,Transcription Factors ,Developmental Biology - Abstract
Pak1 is a member of the PAK family of serine/threonine kinases that are downstream effectors of Rac1 and Cdc42 small GTPases and are implicated in cytoskeleton reorganization. Early expression of Pak1 in Xenopus embryos is tissue restricted, suggesting a role in organogenesis and in cranial neural crest (CNC) cell migration. By observing CNC in vivo and after transplantation, we show that a dominant-negative (DN) Pak1 inhibits its migration. DN-Pak1 also specifically modified the expression of several NC markers. Twist expression was decreased and Snail1 expression posteriorized, but Snail2 (Slug), Sox9 and AP2 were unaffected. DN-Pak1 inhibition of CNC migration could be rescued with Snail1 but not with Twist, which, in fact, cooperated with DN-Pak1 in inhibiting migration. The data confirm that neither Snail1 nor Snail2 expression alone is sufficient for Xenopus CNC migration. Furthermore, they show that, in this tissue, Snail1 and Snail2 expression is not interdependent, nor are these factors subject to obligatory co-regulation, and that their expression depends on signal transduction. Our results also represent the first evidence that Pak1 links extracellular signals to the genetic cascade of transcription factors necessary for CNC specification.
- Published
- 2012
- Full Text
- View/download PDF
35. DNA methyltransferase inhibition may limit cancer cell growth by disrupting ribosome biogenesis
- Author
-
Tom Moss
- Subjects
Cancer Research ,Bisulfite sequencing ,Biology ,Decitabine ,DNA methyltransferase ,Genomic Imprinting ,Epigenetics of physical exercise ,Neoplasms ,Histone methylation ,Humans ,Genes, Tumor Suppressor ,Point-of-View ,DNA (Cytosine-5-)-Methyltransferases ,Gene Silencing ,Cancer epigenetics ,Epigenetics ,Enzyme Inhibitors ,Molecular Biology ,RNA-Directed DNA Methylation ,Cell Proliferation ,Repetitive Sequences, Nucleic Acid ,Genes, rRNA ,DNA Methylation ,HCT116 Cells ,DNA methylation ,Azacitidine ,Cancer research ,CpG Islands ,Ribosomes - Abstract
"Mutations" in the pattern of CpG methylation imprinting of the human genome have been correlated with a number of diseases including cancer. In particular, aberrant imprinting of tumor suppressor genes by gain of CpG methylation has been observed in many cancers and thus represents an important alternative pathway to gene "mutation" and tumor progression. Inhibitors of DNA methylation display therapeutic effects in the treatment of certain cancers, and it has been assumed these effects are due to the reversal of "mutant" gene imprinting. However, significant reactivation of imprinted tumor suppressor genes is rarely observed in vivo following treatment with DNA methylation inhibitors. A recent study revealed an unexpected requirement for CpG methylation in the synthesis and assembly of the ribosome, an essential function for cell growth and proliferation. As such, the data provide an unforeseen explanation of the action of DNA methylation inhibitors in restricting cancer cell growth.
- Published
- 2011
- Full Text
- View/download PDF
36. Extended-Synaptotagmin-2 Mediates FGF Receptor Endocytosis and ERK Activation In Vivo
- Author
-
Michel G. Tremblay, Sabrina Bellenfant, Joëlle Baril, François Guillou, Steve Jean, Tom Moss, and Alexander Mikryukov
- Subjects
MAPK/ERK pathway ,Embryo, Nonmammalian ,Blotting, Western ,Endocytic cycle ,Fluorescent Antibody Technique ,Xenopus Proteins ,Biology ,Fibroblast growth factor ,Endocytosis ,General Biochemistry, Genetics and Molecular Biology ,Mesoderm ,Xbra ,Xenopus laevis ,Adaptor Protein Complex alpha Subunits ,Cell surface receptor ,Synaptotagmin II ,Animals ,Humans ,Immunoprecipitation ,RNA, Messenger ,Receptor, Fibroblast Growth Factor, Type 1 ,Extracellular Signal-Regulated MAP Kinases ,Receptor ,Molecular Biology ,In Situ Hybridization ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Biology ,Cell biology ,Enzyme Activation ,ras Proteins ,Signal transduction ,T-Box Domain Proteins ,Developmental Biology - Abstract
SummaryTargeting of activated plasma membrane receptors to endocytic pathways is important in determining the outcome of growth factor signaling. However, the molecular mechanisms are still poorly understood. Here, we show that the synaptotagmin-related membrane protein E-Syt2 is essential for rapid endocytosis of the activated FGF receptor and for functional signal transduction during Xenopus development. E-Syt2 depletion prevents an early phase of activated FGF receptor endocytosis that we show is required for ERK activation and the induction of the mesoderm. E-Syt2 interacts selectively with the activated FGF receptor and with Adaptin-2, and is required upstream of Ras activation and of receptor autophosphorylation for ERK activation and the induction of the mesodermal marker Xbra. The data identify E-Syt2 as an endocytic adaptor for the clathrin-mediated pathway whose function is conserved in human and suggest a broader role for the E-Syt subfamily in growth factor signaling.
- Published
- 2010
- Full Text
- View/download PDF
37. The ARF Tumor Suppressor Controls Ribosome Biogenesis by Regulating the RNA Polymerase I Transcription Factor TTF-I
- Author
-
Françoise Morin, Stacey Ivanchuk, James T. Rutka, Frédéric Lessard, Tom Moss, Frédéric Langlois, and Victor Y. Stefanovsky
- Subjects
endocrine system ,Nucleolus ,5.8S ribosomal RNA ,Ribosome biogenesis ,RNA-dependent RNA polymerase ,Biology ,Cell Line ,Mice ,03 medical and health sciences ,0302 clinical medicine ,RNA Polymerase I ,Tumor Suppressor Protein p14ARF ,RNA polymerase I ,Animals ,Humans ,Molecular Biology ,Transcription factor ,030304 developmental biology ,0303 health sciences ,Nucleoplasm ,RNA ,Cell Biology ,respiratory system ,Molecular biology ,DNA-Binding Proteins ,Mice, Inbred C57BL ,030220 oncology & carcinogenesis ,NIH 3T3 Cells ,Ribosomes ,HeLa Cells ,Transcription Factors - Abstract
The p14/p19(ARF) (ARF) product of the CDKN2A gene displays tumor suppressor activity both in the presence and absence of p53/TP53. In p53-negative cells, ARF arrests cell proliferation, at least in part, by suppressing ribosomal RNA synthesis. We show that ARF does this by controlling the subnuclear localization of the RNA polymerase I transcription termination factor, TTF-I. TTF-I shuttles between nucleoplasm and nucleolus with the aid of the chaperone NPM/B23 and a nucleolar localization sequence within its N-terminal regulatory domain. ARF inhibits nucleolar import of TTF-I by binding to this nucleolar localization sequence, causing the accumulation of TTF-I in the nucleoplasm. Depletion of TTF-I recapitulates the effects of ARF on ribosomal RNA synthesis and is rescued by the introduction of a TTF-I transgene. Thus, our data delineate the pathway by which ARF regulates ribosomal RNA synthesis and provide a compelling explanation for the role of NPM.
- Published
- 2010
- Full Text
- View/download PDF
38. The splice variants of UBF differentially regulate RNA polymerase I transcription elongation in response to ERK phosphorylation
- Author
-
Tom Moss and Victor Y. Stefanovsky
- Subjects
MAPK/ERK pathway ,Transcription, Genetic ,RNA Splicing ,Biology ,Gene Regulation, Chromatin and Epigenetics ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,RNA Polymerase I ,HMGB Proteins ,Genetics ,RNA polymerase I ,Animals ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Gene ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Genetic Variation ,DNA ,Molecular biology ,Chromatin ,Rats ,chemistry ,Pol1 Transcription Initiation Complex Proteins ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,HMG-Box Domains ,RNA splicing - Abstract
The mammalian architectural HMGB-Box transcription factor UBF is ubiquitously expressed in two variant forms as the result of a differential splicing event, that in the UBF2 deletes 37 amino acid from the second of six HMGB-boxes. Several attempts to define a function for this shorter UBF2 protein have been less than satisfactory. However, since all mammals appear to display similar levels of the longer and shorter UBF variants, it is unlikely that UBF2 is simply nonfunctional. Previously we showed that phosphorylation of UBF by the MAP-kinase ERK regulates chromatin folding and transcription elongation, explaining the rapid response of the ribosomal RNA genes to growth factors. Here we have investigated the roles the UBF variants play in the response of these genes to ERK activity. We demonstrate that the variant HMGB-box 2 of UBF2 has lost the ability to bind bent DNA and hence to induce chromatin folding. As a result it is significantly less effective than UBF1 at arresting RNAPI elongation but at the same time is more responsive to ERK phosphorylation. Thus, UBF2 functionally simulates a hemi-phosphorylated UBF whose expression may provide a means by which to tune the response of the ribosomal RNA genes to growth factor stimulation.
- Published
- 2008
39. Mice lacking both mixed-lineage kinase genes Mlk1 and Mlk2 retain a wild type phenotype
- Author
-
Tom Moss, Steven P. Trusko, Fiona Robinson, Michel J. Tremblay, Nicolas Bisson, and David L. Kaplan
- Subjects
Blotting, Western ,Apoptosis ,Superior Cervical Ganglion ,MAP2K7 ,Mice ,Animals ,ASK1 ,Gene Silencing ,c-Raf ,Muscle, Skeletal ,Molecular Biology ,DNA Primers ,MAPK14 ,Mice, Knockout ,Neurons ,biology ,MAP kinase kinase kinase ,Reverse Transcriptase Polymerase Chain Reaction ,Cyclin-dependent kinase 4 ,Stomach ,Cyclin-dependent kinase 2 ,Cell Biology ,MAP Kinase Kinase Kinases ,Molecular biology ,Cell biology ,Phenotype ,Gastric Mucosa ,biology.protein ,Cyclin-dependent kinase 9 ,Spleen ,Developmental Biology - Abstract
The mitogen-activated protein kinase kinase kinases of the mixed-lineage kinase (MLK) family have been shown to activate the c-Jun N-terminal kinase (JNK) mitogen-activated protein kinase (MAPK) pathway, and to regulate the other two principal MAPK cascades, p38 and extracellular signal-regulated kinase (ERK). Although there is growing evidence for their involvement in neuronal cell death leading to neurodegenerative disorders, little in vivo data is available for the members of this family of kinases. Here, we report that the inactivation of mouse Mlk1 and Mlk2 genes. Mlk1(-/-) and Mlk2(-/-) mice were found to be viable and healthy. Surprisingly, mice carrying the compound Mlk1/Mlk2 null mutations were also found to be viable, fertile and to have a normal life span. The nervous system, testis and kidney, the major sites of MLK1 and 2 expression, all appear normal, as do other organs where these kinases were found to be more weakly expressed. Surprisingly, developmental neuronal programmed cell death, another potential target for MLK family members, was also found to be unaffected. Our results suggest that there is extensive functional redundancy between MLK1/MLK2 and the other member of the family, MLK3, which is also not required for survival in mouse.
- Published
- 2008
- Full Text
- View/download PDF
40. In Vitro DNase I Footprinting
- Author
-
Benoît P, Leblanc and Tom, Moss
- Subjects
DNA-Binding Proteins ,Binding Sites ,DNA Footprinting ,Deoxyribonuclease I ,Electrophoresis, Polyacrylamide Gel ,DNA - Abstract
The association of proteins with the DNA double helix can interfere with the accessibility of the latter to nucleases. This is particularly true when using bulky nucleases such as DNase I. The DNase I footprinting method was developed to take advantage of this fact in the study of DNA-protein interactions: it consists in comparing the pattern of fragments generated by the partial digestion of a DNA sequence in the absence of a protein to that produced by its partial digestion in the presence of said protein. Normally, when the two sets of fragments are separated side by side on a gel, the ladder of DNase I-generated fragments produced in the presence of the protein will feature blank regions (devoid of fragments, indicating protection) and/or enhanced cleavage sites (indicating increased availability to the nuclease). This technique can furthermore reveal if multiple sites for a DNA-binding protein are present on a same fragment and in such a case will also allow the comparison of their respective affinities.
- Published
- 2015
41. The Cruciform DNA Mobility Shift Assay: A Tool to Study Proteins That Recognize Bent DNA
- Author
-
Victor Y, Stefanovsky and Tom, Moss
- Subjects
DNA-Binding Proteins ,DNA, Cruciform ,Binding Sites ,Staining and Labeling ,Antibody Specificity ,Oligonucleotides ,Proteins ,Nucleic Acid Conformation ,Electrophoretic Mobility Shift Assay ,Substrate Specificity ,Protein Binding - Abstract
So-called architectural DNA-binding proteins such as those of the HMGB-box family induce DNA bending and kinking. However, these proteins often display only a weak sequence preference, making the analysis of their DNA-binding characteristics difficult if not impossible in a standard electrophoretic mobility shift assay (EMSA). In contrast, such proteins often bind prebent DNAs with high affinity and specificity. A synthetic cruciform DNA structure will often provide an ideal binding site for such proteins, allowing their affinities for both bent and linear DNAs to be directly and simply determined by a modified form of EMSA.
- Published
- 2015
42. In Vitro DNase I Footprinting
- Author
-
Benoit Leblanc and Tom Moss
- Subjects
Biochemistry ,Protein footprinting ,DNase-I Footprinting ,DNA footprinting ,DNase I hypersensitive site ,Biology ,DNase footprinting assay ,Deoxyribonuclease I ,Hypersensitive site ,Molecular biology ,Footprinting - Abstract
The association of proteins with the DNA double helix can interfere with the accessibility of the latter to nucleases. This is particularly true when using bulky nucleases such as DNase I. The DNase I footprinting method was developed to take advantage of this fact in the study of DNA-protein interactions: it consists in comparing the pattern of fragments generated by the partial digestion of a DNA sequence in the absence of a protein to that produced by its partial digestion in the presence of said protein. Normally, when the two sets of fragments are separated side by side on a gel, the ladder of DNase I-generated fragments produced in the presence of the protein will feature blank regions (devoid of fragments, indicating protection) and/or enhanced cleavage sites (indicating increased availability to the nuclease). This technique can furthermore reveal if multiple sites for a DNA-binding protein are present on a same fragment and in such a case will also allow the comparison of their respective affinities.
- Published
- 2015
- Full Text
- View/download PDF
43. The Cruciform DNA Mobility Shift Assay: A Tool to Study Proteins That Recognize Bent DNA
- Author
-
Tom Moss and Victor Y. Stefanovsky
- Subjects
DNA binding site ,chemistry.chemical_compound ,Biochemistry ,chemistry ,Cruciform ,HMG-box ,Electrophoretic mobility shift assay ,Plasma protein binding ,Binding site ,Biology ,DNA-binding protein ,DNA - Abstract
So-called architectural DNA-binding proteins such as those of the HMGB-box family induce DNA bending and kinking. However, these proteins often display only a weak sequence preference, making the analysis of their DNA-binding characteristics difficult if not impossible in a standard electrophoretic mobility shift assay (EMSA). In contrast, such proteins often bind prebent DNAs with high affinity and specificity. A synthetic cruciform DNA structure will often provide an ideal binding site for such proteins, allowing their affinities for both bent and linear DNAs to be directly and simply determined by a modified form of EMSA.
- Published
- 2015
- Full Text
- View/download PDF
44. Regulation of rRNA Synthesis in Human and Mouse Cells is Not Determined by Changes in Active Gene Count
- Author
-
Tom Moss and Victor Y. Stefanovsky
- Subjects
Transcriptional Activation ,MAPK/ERK pathway ,Gene Dosage ,Biology ,Hydroxamic Acids ,Mice ,Transcription (biology) ,Tumor Cells, Cultured ,Animals ,Humans ,Molecular Biology ,Gene ,Regulation of gene expression ,Epidermal Growth Factor ,Acetylation ,Cell Biology ,Ribosomal RNA ,Molecular biology ,RRNA transcription ,Chromatin ,Enzyme Activation ,Gene Expression Regulation ,RNA, Ribosomal ,Acetyltransferase ,NIH 3T3 Cells ,raf Kinases ,Developmental Biology - Abstract
Growth regulation of the tandemly repeated ribosomal RNA (rRNA) genes in mammals can potentially occur by several distinct mechanisms. Only a fraction of the 200 or so rRNA genes appears to be activated in somatic cells, leaving open the possibility that enhanced transcription could result from gene activation events. Here we have determined the active rRNA gene count after growth stimulation with EGF, direct Raf activation and chromatin hyperacetylation and after inhibiting MAP-kinase signaling. Despite robust changes in rRNA transcription rates, we find no significant variation in active gene number in either mouse fibroblasts or human neuroepithelioma cells. Interestingly, the data also show that rRNA transcription enhancement induced by hyperacetylation is dependent on MEK/ERK signaling. Since ERK and the acetyltransferase CBP both bind the architectural factor UBF, this suggests a mechanism for targeting active CBP to the rRNA genes.
- Published
- 2006
- Full Text
- View/download PDF
45. Growth Factor Signaling Regulates Elongation of RNA Polymerase I Transcription in Mammals via UBF Phosphorylation and r-Chromatin Remodeling
- Author
-
Lawrence I. Rothblum, Frédéric Langlois, Thérèse Gagnon-Kugler, Tom Moss, and Victor Y. Stefanovsky
- Subjects
Transcription, Genetic ,RNA polymerase II ,DNA, Ribosomal ,RNA polymerase III ,Mice ,RNA Polymerase I ,Sigma factor ,RNA polymerase I ,Animals ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Growth Substances ,Molecular Biology ,RNA polymerase II holoenzyme ,General transcription factor ,biology ,Cell Biology ,Chromatin Assembly and Disassembly ,Molecular biology ,RRNA transcription ,RNA, Ribosomal ,NIH 3T3 Cells ,biology.protein ,Transcription factor II D ,Pol1 Transcription Initiation Complex Proteins ,Signal Transduction - Abstract
Synthesis of the 45S rRNA by RNA polymerase I limits cell growth. Knowledge of the mechanism of its regulation is therefore key to understanding growth control. rRNA transcription is believed to be regulated solely at initiation/promoter release. However, we found that stimulation of endogenous 45S rRNA synthesis by epidermal growth factor (EGF) and serum failed to induce an increase in RNA polymerase I engagement on the rRNA genes, despite robust enhancement of 45S rRNA synthesis. Further, endogenous transcription elongation rates were measured and found to be directly proportional to 45S rRNA synthesis. Thus, elongation is a rate-limiting step for rRNA synthesis in vivo. ERK phosphorylation of the HMG boxes of UBF, an RNA polymerase I factor essential for transcription enhancement, was shown to directly regulate elongation by inducing the remodeling of ribosomal gene chromatin. The data suggest a mechanism for coordinating the cotranscriptional assembly of preribosomal particles.
- Published
- 2006
- Full Text
- View/download PDF
46. ERK Modulates DNA Bending and Enhancesome Structure by Phosphorylating HMG1-Boxes 1 and 2 of the RNA Polymerase I Transcription Factor UBF
- Author
-
David P. Bazett-Jones, Victor Y. Stefanovsky, Frédéric Langlois, Tom Moss, and Guillaume Pelletier
- Subjects
MAPK/ERK pathway ,Transcription, Genetic ,HMG-box ,Molecular Sequence Data ,Biology ,Models, Biological ,Biochemistry ,Structure-Activity Relationship ,Xenopus laevis ,chemistry.chemical_compound ,RNA Polymerase I ,Transcription (biology) ,RNA polymerase I ,Animals ,HMGB2 Protein ,Nucleosome ,Amino Acid Sequence ,HMGB1 Protein ,Phosphorylation ,Mitogen-Activated Protein Kinase Kinases ,DNA, Cruciform ,Binding Sites ,DNA ,DNA-binding domain ,Molecular biology ,Recombinant Proteins ,Chromatin ,Enhancer Elements, Genetic ,chemistry ,Mutation ,Pol1 Transcription Initiation Complex Proteins ,Protein Binding - Abstract
Transcription of the ribosomal RNA genes of mammals by RNA polymerase I is rapidly activated by epidermal growth factor via the MAP-kinase (ERK) signaling cascade. This activation is mediated by direct phosphorylation of the HMG box DNA binding domains of the architectural transcription factor UBF. Mutation of the ERK sites of UBF inhibits its normal function and blocks growth factor activation of ribosomal transcription. UBF has little or no DNA sequence selectivity and binds throughout the ribosomal genes, defining a specialized chromatin. Indeed, the HMG boxes of UBF induce looping of the ribosomal DNA to create the enhancesome, a structure somewhat reminiscent of the nucleosome. Here, we show that both ERK phosphorylation and mutations that simulate this phosphorylation decrease the affinity of the individual HMG boxes of UBF for linear ribosomal DNA but have little or no effect on the capacity of these HMG boxes to bind to pre-bent DNA and do not affect the overall binding constant of UBF for the DNA. Electron spectroscopic imaging showed that ERK site UBF mutants do not induce the characteristic DNA looping of the enhancesome and associate with no more than half of the enhancesomal DNA. The data demonstrate that ERK phosphorylation of UBF prevents DNA bending by its first two HMG boxes, leading to a cooperative unfolding of the enhancesome.
- Published
- 2006
- Full Text
- View/download PDF
47. mTOR-Dependent Regulation of Ribosomal Gene Transcription Requires S6K1 and Is Mediated by Phosphorylation of the Carboxy-Terminal Activation Domain of the Nucleolar Transcription Factor UBF†
- Author
-
Alice H. Cavanaugh, Anna Jenkins, Yves Brandenburger, Gretchen Poortinga, Lawrence I. Rothblum, Tom Moss, Katherine M. Hannan, Grant A. McArthur, Ross D. Hannan, Richard B. Pearson, and Kerith Sharkey
- Subjects
Transcription, Genetic ,Ribosome biogenesis ,P70-S6 Kinase 1 ,Biology ,DNA, Ribosomal ,Mice ,Transcription (biology) ,Animals ,Phosphorylation ,Cell Growth and Development ,Molecular Biology ,Transcription factor ,PI3K/AKT/mTOR pathway ,Sirolimus ,Ribosomal Protein S6 Kinases ,TOR Serine-Threonine Kinases ,Cell Biology ,Molecular biology ,Recombinant Proteins ,Protein Structure, Tertiary ,Cell biology ,Enzyme Activation ,Pol1 Transcription Initiation Complex Proteins ,Ribosomal protein s6 ,NIH 3T3 Cells ,Protein Kinases ,Cell Division ,Signal Transduction - Abstract
Mammalian target of rapamycin (mTOR) is a key regulator of cell growth acting via two independent targets, ribosomal protein S6 kinase 1 (S6K1) and 4EBP1. While each is known to regulate translational efficiency, the mechanism by which they control cell growth remains unclear. In addition to increased initiation of translation, the accelerated synthesis and accumulation of ribosomes are fundamental for efficient cell growth and proliferation. Using the mTOR inhibitor rapamycin, we show that mTOR is required for the rapid and sustained serum-induced activation of 45S ribosomal gene transcription (rDNA transcription), a major rate-limiting step in ribosome biogenesis and cellular growth. Expression of a constitutively active, rapamycin-insensitive mutant of S6K1 stimulated rDNA transcription in the absence of serum and rescued rapamycin repression of rDNA transcription. Moreover, overexpression of a dominant-negative S6K1 mutant repressed transcription in exponentially growing NIH 3T3 cells. Rapamycin treatment led to a rapid dephosphorylation of the carboxy-terminal activation domain of the rDNA transcription factor, UBF, which significantly reduced its ability to associate with the basal rDNA transcription factor SL-1. Rapamycin-mediated repression of rDNA transcription was rescued by purified recombinant phosphorylated UBF and endogenous UBF from exponentially growing NIH 3T3 cells but not by hypophosphorylated UBF from cells treated with rapamycin or dephosphorylated recombinant UBF. Thus, mTOR plays a critical role in the regulation of ribosome biogenesis via a mechanism that requires S6K1 activation and phosphorylation of UBF.
- Published
- 2003
- Full Text
- View/download PDF
48. A tissue restricted role for the Xenopus Jun N-terminal kinase kinase kinase MLK2 in cement gland and pronephric tubule differentiation
- Author
-
Tom Moss, Luc Poitras, Nicolas Bisson, and Nazrul Islam
- Subjects
Cellular differentiation ,Xenopus ,Molecular Sequence Data ,Cement gland ,Apoptosis ,Xenopus Proteins ,Biology ,Signal transduction ,Kidney ,Pronephros ,MAP2K7 ,Mixed lineage kinase 2 ,Xenopus laevis ,03 medical and health sciences ,0302 clinical medicine ,GTP-Binding Proteins ,Animals ,ASK1 ,Amino Acid Sequence ,Protein kinase A ,(MLK2, MST, MAP3K10) ,Molecular Biology ,030304 developmental biology ,Programmed cell death ,Leucine Zippers ,0303 health sciences ,Binding Sites ,MAP kinase kinase kinase ,Kinase ,Cell Differentiation ,Cell Biology ,MAP Kinase Kinase Kinases ,biology.organism_classification ,Molecular biology ,030220 oncology & carcinogenesis ,COS Cells ,JNK/SAPK1 ,Developmental Biology - Abstract
The MLK family of mitogen activated protein kinase kinase kinases (MAPKKK) has been shown to activate Jun N-terminal kinase/stress-activated protein kinase 1 (JNK/SAPK1). However, little is known of the in vivo functions of the MLKs. We have identified a Xenopus laevis MLK that shows highest homology with mammalian MLK2 (62%) and, like MLK2, interacts preferentially with the Rho-family GTPase Rac. xMLK2 was expressed zygotically from late gastrula/early neurula. Surprisingly, this expression was restricted to the cement gland, the brain, and the pronephros. In the differentiating cement gland, xMLK2 expression correlated with cell elongation and the onset of a previously unobserved apoptotic phase, while in the pronephros, expression corresponded with the differentiation and opening of the nephric tubules. Overexpression of xMLK2 in COS7 cells led to a SEK1/MKK4 (MAPKK)-dependent hyperactivation of JNK in response to UV irradiation. xMLK2 was shown to be required for normal cement gland development and pronephric tubule formation using antisense inactivation and a dominant negative xMLK2. The data suggest a novel role for the MLKs as tissue-restricted mediators of signal transduction. They also suggest that tissue-specific responses to common extracellular signals may in part result from the programmed expression of MAPKKKs with differing specificities.
- Published
- 2003
- Full Text
- View/download PDF
49. At the Center of Eukaryotic Life
- Author
-
Victor Y. Stefanovsky and Tom Moss
- Subjects
Genetics ,Transcription, Genetic ,Eukaryotic Large Ribosomal Subunit ,Biochemistry, Genetics and Molecular Biology(all) ,5.8S ribosomal RNA ,Computational biology ,Ribosomal RNA ,Biology ,Ribosome ,General Biochemistry, Genetics and Molecular Biology ,Eukaryotic Cells ,Gene Expression Regulation ,Genes ,RNA Polymerase I ,RNA, Ribosomal ,eIF4A ,Animals ,Humans ,Eukaryotic Small Ribosomal Subunit ,30S ,Gene Silencing ,Promoter Regions, Genetic ,Eukaryotic Ribosome ,Cell Nucleolus - Abstract
The ribosomal RNA genes encode the enzymatic scaffold of the ribosome and thereby perform perhaps the most basic of all housekeeping functions. However, recent data suggests that they might also control important aspects of cell behavior.
- Published
- 2002
- Full Text
- View/download PDF
50. An Immediate Response of Ribosomal Transcription to Growth Factor Stimulation in Mammals Is Mediated by ERK Phosphorylation of UBF
- Author
-
Ross D. Hannan, Guillaume Pelletier, Victor Y. Stefanovsky, Lawrence I. Rothblum, Tom Moss, and Thérèse Gagnon-Kugler
- Subjects
Transcription, Genetic ,Recombinant Fusion Proteins ,Response element ,E-box ,Protein Structure, Secondary ,Mice ,Sp3 transcription factor ,Genes, Reporter ,RNA Polymerase I ,Serum response factor ,Animals ,Humans ,Phosphorylation ,Molecular Biology ,Mitogen-Activated Protein Kinase 1 ,Sp1 transcription factor ,Mitogen-Activated Protein Kinase 3 ,Epidermal Growth Factor ,biology ,General transcription factor ,DNA ,Cell Biology ,Molecular biology ,Activating transcription factor 2 ,DNA-Binding Proteins ,Enzyme Activation ,RNA, Ribosomal ,Mutation ,biology.protein ,Mitogen-Activated Protein Kinases ,Pol1 Transcription Initiation Complex Proteins ,Ribosomes ,Transcription factor II B ,Transcription Factors - Abstract
Ribosomal transcription in mammals is regulated in response to growth, differentiation, disease, and aging, but the mechanisms of this regulation have remained unresolved. We show that epidermal growth factor induces immediate, ERK1/2-dependent activation of endogenous ribosomal transcription, while inactivation of ERK1/2 causes an equally immediate reversion to the basal transcription level. ERK1/2 was found to phosphorylate the architectural transcription factor UBF at amino acids 117 and 201 within HMG boxes 1 and 2, preventing their interaction with DNA. Mutation of these sites inhibited transcription activation and abrogated the transcriptional response to ERK1/2. Thus, growth factor regulation of ribosomal transcription likely acts by a cyclic modulation of DNA architecture. The data suggest a central role for ribosome biogenesis in growth regulation.
- Published
- 2001
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.