41 results on '"Elmar Wolf"'
Search Results
2. MondoA drives malignancy in B-ALL through enhanced adaptation to metabolic stress
- Author
-
Andreas Petry, Rupert Öllinger, Thomas G. P. Grunewald, Stefan Burdach, Gaurav Jain, Davide G. Franchina, Erik Hameister, Agnes Görlach, Poul H. Sorensen, Michaela Carina Baldauf, Juliane Schmäh, Jürgen Ruland, Carolin Prexler, Elmar Wolf, Gunther Richter, Dirk Brenner, Alexandra Sipol, Uwe Thiel, Roland Rad, Gunnar Cario, Maxim Barenboim, Julia Hofstetter, Busheng Xue, and Rebeca Alba Rubio
- Subjects
Mice, Knockout ,Mice, Inbred BALB C ,Cell division ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,Immunology ,Cell Biology ,Hematology ,Oxidative phosphorylation ,Biology ,Biochemistry ,Interactome ,Neoplasm Proteins ,Cell biology ,Glutamine ,Mice ,Metabolic pathway ,Stress, Physiological ,RNA interference ,Cell Line, Tumor ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Cancer cell ,Animals ,Humans ,Homeostasis - Abstract
Cancer cells are in most instances characterized by rapid proliferation and uncontrolled cell division. Hence, they must adapt to proliferation-induced metabolic stress through intrinsic or acquired antimetabolic stress responses to maintain homeostasis and survival. One mechanism to achieve this is reprogramming gene expression in a metabolism-dependent manner. MondoA (also known as Myc-associated factor X–like protein X-interacting protein [MLXIP]), a member of the MYC interactome, has been described as an example of such a metabolic sensor. However, the role of MondoA in malignancy is not fully understood and the underlying mechanism in metabolic responses remains elusive. By assessing patient data sets, we found that MondoA overexpression is associated with worse survival in pediatric common acute lymphoblastic leukemia (ALL; B-precursor ALL [B-ALL]). Using clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) and RNA-interference approaches, we observed that MondoA depletion reduces the transformational capacity of B-ALL cells in vitro and dramatically inhibits malignant potential in an in vivo mouse model. Interestingly, reduced expression of MondoA in patient data sets correlated with enrichment in metabolic pathways. The loss of MondoA correlated with increased tricarboxylic acid cycle activity. Mechanistically, MondoA senses metabolic stress in B-ALL cells by restricting oxidative phosphorylation through reduced pyruvate dehydrogenase activity. Glutamine starvation conditions greatly enhance this effect and highlight the inability to mitigate metabolic stress upon loss of MondoA in B-ALL. Our findings give novel insight into the function of MondoA in pediatric B-ALL and support the notion that MondoA inhibition in this entity offers a therapeutic opportunity and should be further explored.
- Published
- 2022
- Full Text
- View/download PDF
3. The glycolytic enzyme ALDOA and the exon junction complex protein RBM8A are regulators of ribosomal biogenesis
- Author
-
Jessica Denise Schwarz, Sören Lukassen, Pranjali Bhandare, Lorenz Eing, Marteinn Thor Snaebjörnsson, Yiliam Cruz García, Jan Philipp Kisker, Almut Schulze, and Elmar Wolf
- Subjects
ddc:570 ,Cell Biology ,Developmental Biology - Abstract
Cellular growth is a fundamental process of life and must be precisely controlled in multicellular organisms. Growth is crucially controlled by the number of functional ribosomes available in cells. The production of new ribosomes depends critically on the activity of RNA polymerase (RNAP) II in addition to the activity of RNAP I and III, which produce ribosomal RNAs. Indeed, the expression of both, ribosomal proteins and proteins required for ribosome assembly (ribosomal biogenesis factors), is considered rate-limiting for ribosome synthesis. Here, we used genetic screening to identify novel transcriptional regulators of cell growth genes by fusing promoters from a ribosomal protein gene (Rpl18) and from a ribosomal biogenesis factor (Fbl) with fluorescent protein genes (RFP, GFP) as reporters. Subsequently, both reporters were stably integrated into immortalized mouse fibroblasts, which were then transduced with a genome-wide sgRNA-CRISPR knockout library. Subsequently, cells with altered reporter activity were isolated by FACS and the causative sgRNAs were identified. Interestingly, we identified two novel regulators of growth genes. Firstly, the exon junction complex protein RBM8A controls transcript levels of the intronless reporters used here. By acute depletion of RBM8A protein using the auxin degron system combined with the genome-wide analysis of nascent transcription, we showed that RBM8A is an important global regulator of ribosomal protein transcripts. Secondly, we unexpectedly observed that the glycolytic enzyme aldolase A (ALDOA) regulates the expression of ribosomal biogenesis factors. Consistent with published observations that a fraction of this protein is located in the nucleus, this may be a mechanism linking transcription of growth genes to metabolic processes and possibly to metabolite availability.
- Published
- 2022
4. The HSV-1 ICP22 protein selectively impairs histone repositioning upon Pol II transcription downstream of genes
- Author
-
Andrea Milic, Katharina Wolf, Elmar Wolf, Katharina Reinisch, Caroline C. Friedel, Michael Kluge, Christopher Juerges, Lars Dölken, Lara Djakovic, Florian Erhard, Thomas Hennig, Adam W. Whisnant, Elena Weiß, Arnhild Grothey, and Tobias Haas
- Subjects
Histone ,biology ,Downstream (manufacturing) ,Chemistry ,Transcription (biology) ,viruses ,biology.protein ,RNA polymerase II ,HSL and HSV ,Gene ,Cell biology - Abstract
Herpes simplex virus 1 (HSV-1) infection and stress responses disrupt transcription termination by RNA Polymerase II (Pol II). In HSV-1 infection but not upon salt or heat stress, this is accompanied by a dramatic increase in chromatin accessibility downstream of genes. Here, we show that the HSV-1 immediate-early protein ICP22 is both necessary and sufficient to induce downstream open chromatin regions (dOCRs) upon disrupted transcription termination. This was matched by a marked ICP22-dependent loss of histones downstream of affected genes consistent with impaired histone repositioning in the wake of Pol II. Efficient knock-down of the ICP22-interacting histone chaperon FACT, though insufficient to induce dOCRs in ΔICP22 infection, increased dOCR induction upon wild-type HSV-1 infection. Interestingly, this was accompanied by a marked increase in chromatin accessibility within gene bodies. We propose a model in which allosteric changes in Pol II composition downstream of genes and ICP22-mediated interference with FACT activity explain the differential impairment of histone re-positioning in the wake of Pol II observed in HSV-1 infection.
- Published
- 2021
- Full Text
- View/download PDF
5. MYC- and MIZ1-Dependent Vesicular Transport of Double-Strand RNA Controls Immune Evasion in Pancreatic Ductal Adenocarcinoma
- Author
-
Lars Zender, Carsten P. Ade, Mathias T. Rosenfeldt, Emilia Vendelova, Ursula Eilers, Bastian Krenz, Elmar Wolf, Anneli Gebhardt-Wolf, Armin Wiegering, Peter Gallant, Abdallah Gaballa, Apoorva Baluapuri, Stefan Bauer, Florian Roehrig, Luana D’Artista, Georg Gasteiger, and Martin Eilers
- Subjects
Cancer Research ,Kruppel-Like Transcription Factors ,Mice, Nude ,Biology ,Adenocarcinoma ,Protein Serine-Threonine Kinases ,Article ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Paracrine signalling ,Mice ,0302 clinical medicine ,Immune system ,TANK-binding kinase 1 ,Mice, Inbred NOD ,MHC class I ,Animals ,Humans ,Autocrine signalling ,030304 developmental biology ,Immune Evasion ,RNA, Double-Stranded ,Cell Nucleus ,0303 health sciences ,Mice, Inbred BALB C ,RNA ,Biological Transport ,Sequence Analysis, DNA ,Introns ,Cell biology ,Mice, Inbred C57BL ,Pancreatic Neoplasms ,RNA silencing ,HEK293 Cells ,Oncology ,030220 oncology & carcinogenesis ,Immune System ,TLR3 ,biology.protein ,Tumor Suppressor Protein p53 ,Gene Deletion ,Carcinoma, Pancreatic Ductal - Abstract
Deregulated expression of the MYC oncoprotein enables tumor cells to evade immune surveillance, but the mechanisms underlying this surveillance are poorly understood. We show here that endogenous MYC protects pancreatic ductal adenocarcinoma (PDAC) driven by KRASG12D and TP53R172H from eradication by the immune system. Deletion of TANK-binding kinase 1 (TBK1) bypassed the requirement for high MYC expression. TBK1 was active due to the accumulation of double-stranded RNA (dsRNA), which was derived from inverted repetitive elements localized in introns of nuclear genes. Nuclear-derived dsRNA is packaged into extracellular vesicles and subsequently recognized by toll-like receptor 3 (TLR3) to activate TBK1 and downstream MHC class I expression in an autocrine or paracrine manner before being degraded in lysosomes. MYC suppressed loading of dsRNA onto TLR3 and its subsequent degradation via association with MIZ1. Collectively, these findings suggest that MYC and MIZ1 suppress a surveillance pathway that signals perturbances in mRNA processing to the immune system, which facilitates immune evasion in PDAC. Significance: This study identifies a TBK1-dependent pathway that links dsRNA metabolism to antitumor immunity and shows that suppression of TBK1 is a critical function of MYC in pancreatic ductal adenocarcinoma.
- Published
- 2021
6. Design, Synthesis and Evaluation of WD-repeat containing protein 5 (WDR5) degraders
- Author
-
N. Berner, Magda Szewczyk, B. Kuester, J. Weckesser, Martin Eilers, Andreas Kraemer, Christoph A. Sotriffer, Volker Doetsch, Bikash Adhikari, Frank Loehr, Stephanie Heinzlmeir, Dalia Barsyte-Lovejoy, A. Doelle, Mathias Diebold, Elmar Wolf, J. Gerbel, Lena M. Berger, Stefan Knapp, and Cheryl H. Arrowsmith
- Subjects
Scaffold protein ,Histone ,biology ,Chemistry ,WD Repeat-Containing Protein 5 ,Histone methyltransferase ,biology.protein ,WDR5 ,Gene ,Transcription factor ,Function (biology) ,Cell biology - Abstract
Histone H3K4 methylation serves as post-translational hallmark of actively transcribed genes and is introduced by histone methyltransferases (HMT) and its regulatory scaffolding proteins. One of these is the WD-repeat containing protein 5 (WDR5) that has also been associated with controlling long non-coding RNAs and transcription factors including MYC. The wide influence of dysfunctional HMTs complexes and the typically upregulated MYC levels in diverse tumor types suggested WDR5 as an attractive drug target. Indeed, protein-protein interface inhibitors for two protein interaction interfaces on WDR5 have been developed. While such compounds only inhibit a subset of WDR5 interactions, chemically induced proteasomal degradation of WDR5 might represent an elegant way to target all oncogenic function. This study presents the design, synthesis and evaluation of two diverse WDR5 degrader series based on two WIN site binding scaffolds and shows that linker nature and length strongly influence degradation efficacy.
- Published
- 2021
- Full Text
- View/download PDF
7. MondoA Drives Malignancy in cALL through Enhanced Adaptation to Metabolic Stress
- Author
-
Elmar Wolf, Rupert Öllinger, Jürgen Ruland, Gaurav Jain, Stefan Burdach, Gunther Richter, Andreas Petry, Michaela C. Baldauf, Uwe Thiel, Carolin Prexler, Poul H. Sorensen, Erik Hameister, Agnes Görlach, Julia Hofstetter, Maxim Barenboim, Thomas G. P. Grunewald, Juliane Schmäh, Busheng Xue, Davide G. Franchina, Rebeca Alba Rubio, Roland Rad, Gunnar Cario, Alexandra Sipol, and Dirk Brenner
- Subjects
Glutamine ,Metabolic pathway ,Cell division ,RNA interference ,Cancer cell ,Oxidative phosphorylation ,Biology ,Interactome ,Homeostasis ,Cell biology - Abstract
Cancer cells are in most instances characterized by rapid proliferation and uncontrolled cell division. Hence, they must adapt to proliferation-induced metabolic stress through intrinsic or acquired anti-metabolic stress responses to maintain homeostasis and survival. One mechanism to achieve this is to reprogram gene expression in a metabolism-dependent manner. MondoA (also known as MLXIP), a member of the MYC interactome, has been described as an example of such a metabolic sensor. However, the role of MondoA in malignancy is not fully understood and the underlying mechanism in metabolic responses remains elusive. By assessing patient data sets we found that MondoA overexpression is associated with a worse survival in pediatric common acute lymphoblastic leukemia (cALL). Using CRISPR/Cas9 and RNA interference approaches, we observed that MondoA depletion reduces transformational capacity of cALL cells in vitro and dramatically inhibits malignant potential in an in vivo mouse model. Interestingly, reduced expression of MondoA in patient data sets correlated with enrichment in metabolic pathways. The loss of MondoA correlated with increased tricarboxylic acid (TCA) cycle activity. Mechanistically, MondoA senses metabolic stress in cALL cells by restricting oxidative phosphorylation through reduced PDH activity. Glutamine starvation conditions greatly enhance this effect and highlight the inability to mitigate metabolic stress upon loss of MondoA in cALL. Our findings give a novel insight into the function of MondoA in pediatric cALL and support the notion that MondoA inhibition in this entity offers a therapeutic opportunity and should be further explored.Key PointsMondoA maintains aggressiveness and leukemic burden in common ALL, modulating metabolic stress response.
- Published
- 2020
- Full Text
- View/download PDF
8. Decision letter: MYC regulates ribosome biogenesis and mitochondrial gene expression programs through its interaction with host cell factor–1
- Author
-
Elmar Wolf and Martin Eilers
- Subjects
Mitochondrial DNA ,medicine.anatomical_structure ,Interaction with host ,Cell ,medicine ,Ribosome biogenesis ,Biology ,Cell biology - Published
- 2020
- Full Text
- View/download PDF
9. Plant roots employ cell-layer-specific programs to respond to pathogenic and beneficial microbes
- Author
-
William Andres Lopez-Arboleda, Jaqueline Komorek, Joëlle Le Berre, Agnès Attard, Alexander Marsell, Elmar Wolf, Niko Geldner, Wolfgang Dröge-Laser, Arthur Korte, Frank Waller, and Christian Fröschel
- Subjects
Phytophthora ,Arabidopsis ,Microbiology ,Endophyte ,Ribosome ,Plant Roots ,03 medical and health sciences ,0302 clinical medicine ,Ascomycota ,Verticillium longisporum ,Gene Expression Regulation, Plant ,Virology ,Plant Immunity ,Symbiosis ,Pathogen ,030304 developmental biology ,Plant Diseases ,Oomycete ,0303 health sciences ,biology ,Basidiomycota ,RNA ,biology.organism_classification ,Isolation (microbiology) ,Cell biology ,Rhizosphere ,Parasitology ,Central cylinder ,030217 neurology & neurosurgery - Abstract
Summary Plant roots are built of concentric cell layers that are thought to respond to microbial infections by employing specific, genetically defined programs. Yet, the functional impact of this radial organization remains elusive, particularly due to the lack of genome-wide techniques for monitoring expression at a cell-layer resolution. Here, cell-type-specific expression of tagged ribosomes enabled the isolation of ribosome-bound mRNA to obtain cell-layer translatomes (TRAP-seq, translating ribosome affinity purification and RNA sequencing). After inoculation with the vascular pathogen Verticillium longisporum, pathogenic oomycete Phytophthora parasitica, or mutualistic endophyte Serendipita indica, root cell-layer responses reflected the fundamentally different colonization strategies of these microbes. Notably, V. longisporum specifically suppressed the endodermal barrier, which restricts fungal progression, allowing microbial access to the root central cylinder. Moreover, localized biosynthesis of antimicrobial compounds and ethylene differed in response to pathogens and mutualists. These examples highlight the power of this resource to gain insights into root-microbe interactions and to develop strategies in crop improvement.
- Published
- 2020
10. PROTAC-mediated degradation reveals a non-catalytic function of AURORA-A kinase
- Author
-
Stephanie Heinzlmeir, Bikash Adhikari, Bernhard Kuster, Ashwin Narain, Pranjali Bhandare, Jessica Denise Schwarz, Lars Schönemann, Mathias Diebold, Nevenka Dudvarski Stankovic, Julia Hofstetter, Lorenz Eing, Martin Schröder, Stefan Knapp, Elmar Wolf, Apoorva Baluapuri, Christoph A. Sotriffer, Jelena Bozilovic, Marek Wanior, Andreas Schlosser, and Markus Vogt
- Subjects
Male ,CEREBLON ,Protein Conformation ,Apoptosis ,Polyethylene Glycols ,Catalytic Domain ,Molecular Targeted Therapy ,Aurora Kinase A ,AURORA-A ,0303 health sciences ,MLN8237 ,Kinase ,Chemistry ,Cell Cycle ,030302 biochemistry & molecular biology ,Cell cycle ,Small molecule ,Cell biology ,embryonic structures ,Female ,biological phenomena, cell phenomena, and immunity ,Protein Binding ,DNA Replication ,Alisertib ,S-phase ,Ubiquitin-Protein Ligases ,Aurora A kinase ,Antineoplastic Agents ,macromolecular substances ,Article ,PROTAC ,03 medical and health sciences ,Cell Line, Tumor ,thalidomide ,Humans ,cancer ,MLN8054 ,Protein Kinase Inhibitors ,Molecular Biology ,Mitosis ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,Cereblon ,DNA replication ,Cooperative binding ,Cell Biology ,Benzazepines ,enzymes and coenzymes (carbohydrates) ,Drug Design ,Degronimid ,Proteolysis ,targeted protein degradation - Abstract
The mitotic kinase AURORA-A is essential for cell cycle progression and is considered a priority cancer target. Although the catalytic activity of AURORA-A is essential for its mitotic function, recent reports indicate an additional non-catalytic function, which is difficult to target by conventional small molecules. We therefore developed a series of chemical degraders (PROTACs) by connecting a clinical kinase inhibitor of AURORA-A to E3 ligase-binding molecules (for example, thalidomide). One degrader induced rapid, durable and highly specific degradation of AURORA-A. In addition, we found that the degrader complex was stabilized by cooperative binding between AURORA-A and CEREBLON. Degrader-mediated AURORA-A depletion caused an S-phase defect, which is not the cell cycle effect observed upon kinase inhibition, supporting an important non-catalytic function of AURORA-A during DNA replication. AURORA-A degradation induced rampant apoptosis in cancer cell lines and thus represents a versatile starting point for developing new therapeutics to counter AURORA-A function in cancer.
- Published
- 2020
- Full Text
- View/download PDF
11. Targeting MYC Proteins for Tumor Therapy
- Author
-
Martin Eilers and Elmar Wolf
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,Tumor therapy ,Cell Biology ,Myc proteins ,3. Good health ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Alisertib ,Cancer research ,Medicine ,WDR5 ,business - Abstract
Targeting the function of MYC oncoproteins holds the promise of achieving conceptually new and effective anticancer therapies that can be applied to a broad range of tumors. The nature of the target however—a broadly, possibly universally acting transcription factor that has no enzymatic activity and is largely unstructured unless complexed with partner proteins—has so far defied the development of clinically applicable MYC-directed therapies. At the same time, lingering questions about exactly which functions of MYC proteins account for their pervasive oncogenic role in human tumors and need to be targeted have prevented the development of effective therapies using surrogate targets that act in critical MYC-dependent pathways. In this review, we therefore argue that rigorous testing of critical oncogenic functions and protein/protein interactions and new chemical approaches to target them are necessary to successfully eradicate MYC-driven tumors.
- Published
- 2020
- Full Text
- View/download PDF
12. Snf1-RELATED KINASE1-Controlled C/S1-bZIP Signaling Activates Alternative Mitochondrial Metabolic Pathways to Ensure Plant Survival in Extended Darkness
- Author
-
Markus Teige, Elmar Wolf, Katrin Dietrich, Thomas Nägele, Andrea Mair, Lorenzo Pedrotti, Christoph Weiste, Francesca Lorenzin, Wolfgang Dröge-Laser, Elena Baena-González, and Wolfram Weckwerth
- Subjects
0106 biological sciences ,0301 basic medicine ,Leucine zipper ,Promoter ,Cell Biology ,Plant Science ,Biology ,01 natural sciences ,Cell biology ,Chromatin ,03 medical and health sciences ,030104 developmental biology ,Histone ,Transcription (biology) ,biology.protein ,Signal transduction ,Gene ,Transcription factor ,010606 plant biology & botany - Abstract
Sustaining energy homeostasis is of pivotal importance for all living organisms. In Arabidopsis thaliana, evolutionarily conserved SnRK1 kinases (Snf1-RELATED KINASE1) control metabolic adaptation during low energy stress. To unravel starvation-induced transcriptional mechanisms, we performed transcriptome studies of inducible knockdown lines and found that S1-basic leucine zipper transcription factors (S1-bZIPs) control a defined subset of genes downstream of SnRK1. For example, S1-bZIPs coordinate the expression of genes involved in branched-chain amino acid catabolism, which constitutes an alternative mitochondrial respiratory pathway that is crucial for plant survival during starvation. Molecular analyses defined S1-bZIPs as SnRK1-dependent regulators that directly control transcription via binding to G-box promoter elements. Moreover, SnRK1 triggers phosphorylation of group C-bZIPs and the formation of C/S1-heterodimers and, thus, the recruitment of SnRK1 directly to target promoters. Subsequently, the C/S1-bZIP-SnRK1 complex interacts with the histone acetylation machinery to remodel chromatin and facilitate transcription. Taken together, this work reveals molecular mechanisms underlying how energy deprivation is transduced to reprogram gene expression, leading to metabolic adaptation upon stress.
- Published
- 2018
- Full Text
- View/download PDF
13. Targeted protein degradation reveals a direct role of SPT6 in RNAPII elongation and termination
- Author
-
Andreas Schlosser, Martin Eilers, Bikash Adhikari, Simone Backes, Pranjali Bhandare, Elmar Wolf, Apoorva Baluapuri, Florian Erhard, Ashwin Narain, and Lars Dölken
- Subjects
DNA Replication ,termination ,Transcription Elongation, Genetic ,elongation ,RNA polymerase II ,Protein degradation ,Polyadenylation ,SUPT6H ,Article ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Transcription (biology) ,Humans ,Epigenetics ,Molecular Biology ,Gene ,030304 developmental biology ,0303 health sciences ,Indoleacetic Acids ,biology ,mathematical modeling ,Cell Biology ,Cell biology ,SPT6 ,Elongation factor ,Histone ,histone chaperone ,Chaperone (protein) ,Proteolysis ,biology.protein ,RNA ,auxin ,transcription ,targeted protein degradation ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Summary SPT6 is a histone chaperone that tightly binds RNA polymerase II (RNAPII) during transcription elongation. However, its primary role in transcription is uncertain. We used targeted protein degradation to rapidly deplete SPT6 in human cells and analyzed defects in RNAPII behavior by a multi-omics approach and mathematical modeling. Our data indicate that SPT6 is a crucial factor for RNAPII processivity and is therefore required for the productive transcription of protein-coding genes. Unexpectedly, SPT6 also has a vital role in RNAPII termination, as acute depletion induced readthrough transcription for thousands of genes. Long-term depletion of SPT6 induced cryptic intragenic transcription, as observed earlier in yeast. However, this phenotype was not observed upon acute SPT6 depletion and therefore can be attributed to accumulated epigenetic perturbations in the prolonged absence of SPT6. In conclusion, targeted degradation of SPT6 allowed the temporal discrimination of its function as an epigenetic safeguard and RNAPII elongation factor., Graphical abstract, Highlights • Auxin-inducible degradation discriminates direct roles of human SPT6 in transcription • Acute loss of SPT6 globally impairs RNAPII processivity and speed • SPT6 is required for efficient transcription termination on protein-coding genes • Long-term loss of SPT6 ultimately results in cryptic intragenic transcription, Histone chaperone SPT6 is essential for maintaining epigenetic integrity in the wake of transcription by RNAP II. By studying the effects of targeted acute depletion of SPT6 with multi-omics and mathematical modeling, Narain et al. uncovered direct roles of SPT6 in ensuring efficient transcription elongation and termination in human cells.
- Published
- 2021
- Full Text
- View/download PDF
14. A central role of glutamine in Chlamydia infection
- Author
-
Nadine Vollmuth, Thomas F. Wulff, Werner Schmitz, Claudia Huber, Julian Fink, Wolfgang Eisenreich, Apoorva Baluapuri, Jürgen Seibel, Francesca R Dejure, Sudha Janaki-Raman, Karthika Rajeeve, Thomas Rudel, Elmar Wolf, Almut Schulze, and Maximilian Schmalhofer
- Subjects
Chlamydia ,Glutaminase ,Intracellular parasite ,Biology ,medicine.disease_cause ,medicine.disease ,Cell biology ,Glutamine ,chemistry.chemical_compound ,chemistry ,medicine ,Peptidoglycan ,Chlamydia trachomatis ,Reprogramming ,Intracellular - Abstract
Obligate intracellular bacteria like Chlamydia trachomatis undergo a complex developmental cycle between infectious non-replicative (EBs) and non-infectious replicative (RBs) forms. EBs shortly after entering a host cell transform to RBs, a crucial process in infection, initiating chlamydial replication. As Chlamydia fail to replicate outside the host cell it is currently unknown how the transition from EBs to RBs is initiated. Here we show in a cell-free approach in axenic media that uptake of glutamine by the bacteria is critical to initiate EB-RB transition. These bacteria utilize glutamine to synthesize cell wall peptidoglycan which has recently been detected in the septa of replicating intracellular Chlamydia. The increased requirement for glutamine in infected cells is achieved by reprogramming the glutamine metabolism in a c-Myc-dependent manner. Glutamine was effectively taken up by the glutamine transporter SLC1A5 and metabolized via glutaminase. Interference with this metabolic reprogramming limited growth of Chlamydia. Intriguingly, Chlamydia failed to produce progeny in SLC1A5 knockout mice. Thus, we report on the central role of glutamine for the development of an obligate intracellular pathogenic bacterium and the reprogramming of host glutamine metabolism, which may provide a basis for innovative anti-infective strategies.
- Published
- 2019
- Full Text
- View/download PDF
15. MYC competes with MiT/TFE in regulating lysosomal biogenesis and autophagy through an epigenetic rheostat
- Author
-
Heather Tillman, Geoffrey Neale, Yvan Campos, Eda Machado, Martine F. Roussel, Rosario Mosca, Leigh E. Fremuth, Elmar Wolf, Gerard Grosveld, Ida Annunziata, Diantha van de Vlekkert, David Finkelstein, Alessandra d'Azzo, Xiaohui Qiu, Min-Joon Han, and Jason A. Weesner
- Subjects
0301 basic medicine ,Transcription, Genetic ,General Physics and Astronomy ,Histone Deacetylase 2 ,02 engineering and technology ,Epigenesis, Genetic ,Induced pluripotent stem cell ,lcsh:Science ,Promoter Regions, Genetic ,Polytetrafluoroethylene ,Cancer ,Regulation of gene expression ,Multidisciplinary ,Organelle Biogenesis ,biology ,Chemistry ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,Stem Cells ,Forkhead Transcription Factors ,021001 nanoscience & nanotechnology ,Cancer metabolism ,3. Good health ,Cell biology ,Gene Expression Regulation, Neoplastic ,Histone ,Mechanisms of disease ,Colonic Neoplasms ,0210 nano-technology ,Science ,General Biochemistry, Genetics and Molecular Biology ,Article ,Histone Deacetylases ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Cell Line, Tumor ,Autophagy ,Humans ,Epigenetics ,Transcription factor ,Binding Sites ,General Chemistry ,030104 developmental biology ,biology.protein ,TFEB ,lcsh:Q ,Organelle biogenesis ,Lysosomes - Abstract
Coordinated regulation of the lysosomal and autophagic systems ensures basal catabolism and normal cell physiology, and failure of either system causes disease. Here we describe an epigenetic rheostat orchestrated by c-MYC and histone deacetylases that inhibits lysosomal and autophagic biogenesis by concomitantly repressing the expression of the transcription factors MiT/TFE and FOXH1, and that of lysosomal and autophagy genes. Inhibition of histone deacetylases abates c-MYC binding to the promoters of lysosomal and autophagy genes, granting promoter occupancy to the MiT/TFE members, TFEB and TFE3, and/or the autophagy regulator FOXH1. In pluripotent stem cells and cancer, suppression of lysosomal and autophagic function is directly downstream of c-MYC overexpression and may represent a hallmark of malignant transformation. We propose that, by determining the fate of these catabolic systems, this hierarchical switch regulates the adaptive response of cells to pathological and physiological cues that could be exploited therapeutically., Genes related to lysosomal and autophagic systems are transcriptionally regulated by the Mit/TFE family of transcription factors. Here the authors show that MYC, in association with HDACs, suppresses the expression of lysosomal and autophagy genes by competing with the Mit/TFE transcription factors for occupancy of their target gene promoters.
- Published
- 2019
16. Reprogramming of host glutamine metabolism during Chlamydia trachomatis infection and its key role in peptidoglycan synthesis
- Author
-
Julian Fink, Rajeeve Sivadasan, Jürgen Seibel, Martin Eilers, Maximilian Schmalhofer, Sudha Janaki-Raman, Apoorva Baluapuri, Nadine Vollmuth, Thomas F. Wulff, Thomas Rudel, Werner Schmitz, Francesca R Dejure, Claudia Huber, Karthika Rajeeve, Elmar Wolf, Almut Schulze, Wolfgang Eisenreich, and HIRI, Helmholtz-Institut für RNA-basierte Infektionsforschung, Josef-Shneider Strasse 2, 97080 Würzburg, Germany.
- Subjects
Microbiology (medical) ,EXPRESSION ,Amino Acid Transport System ASC ,Glutamine ,Immunology ,Chlamydia trachomatis ,Peptidoglycan ,Biology ,medicine.disease_cause ,Applied Microbiology and Biotechnology ,Microbiology ,Cell Line ,ACTIVATION ,Minor Histocompatibility Antigens ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Mice ,Phosphatidylinositol 3-Kinases ,BINDING ,Genetics ,medicine ,C-MYC ,Animals ,Humans ,CYCLE ,030304 developmental biology ,Mitogen-Activated Protein Kinase Kinases ,0303 health sciences ,Chlamydia ,Obligate ,030306 microbiology ,Glutaminase ,Intracellular parasite ,GLUCOSE-METABOLISM ,Cell Biology ,Chlamydia Infections ,medicine.disease ,3. Good health ,Cell biology ,CHLAMYDIA-TRACHOMATIS ,Gene Expression Regulation ,CELLS ,Host-Pathogen Interactions ,GROWTH ,Signal transduction ,Reprogramming ,Signal Transduction - Abstract
Obligate intracellular bacteria such as Chlamydia trachomatis undergo a complex developmental cycle between infectious, non-replicative elementary-body and non-infectious, replicative reticulate-body forms. Elementary bodies transform to reticulate bodies shortly after entering a host cell, a crucial process in infection, initiating chlamydial replication. As Chlamydia fail to replicate outside the host cell, it is unknown how the replicative part of the developmental cycle is initiated. Here we show, using a cell-free approach in axenic media, that the uptake of glutamine by the bacteria is crucial for peptidoglycan synthesis, which has a role in Chlamydia replication. The increased requirement for glutamine in infected cells is satisfied by reprogramming the glutamine metabolism in a c-Myc-dependent manner. Glutamine is effectively taken up by the glutamine transporter SLC1A5 and metabolized via glutaminase. Interference with this metabolic reprogramming limits the growth of Chlamydia. Intriguingly, Chlamydia failed to produce progeny in SLC1A5-knockout organoids and mice. Thus, we report on the central role of glutamine for the development of an obligate intracellular pathogenic bacterium and the reprogramming of host glutamine metabolism, which may provide a basis for innovative anti-infection strategies.
- Published
- 2019
- Full Text
- View/download PDF
17. Recruitment of BRCA1 limits MYCN-driven accumulation of stalled RNA polymerase
- Author
-
Elmar Wolf, Martin Eilers, Jan Koster, Apoorva Baluapuri, Rogier Versteeg, Sabrina Rodewald, Daniel Solvie, Gabriele Büchel, Jacqueline Kalb, Carsten P. Ade, Jiajia Xu, Steffi Herold, Jan J. Molenaar, Anne Carstensen, Susanne Walz, Matthias Dobbelstein, Christina Klotz, Christina Schülein-Völk, Oncogenomics, and CCA - Cancer biology and immunology
- Subjects
Transcription Elongation, Genetic ,RNA polymerase II ,Article ,Neuroblastoma ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Line, Tumor ,RNA polymerase ,medicine ,Humans ,Transcription factor ,neoplasms ,030304 developmental biology ,Regulation of gene expression ,N-Myc Proto-Oncogene Protein ,0303 health sciences ,Messenger RNA ,Multidisciplinary ,biology ,BRCA1 Protein ,Protein Stability ,Promoter ,medicine.disease ,Chromatin ,Cell biology ,Gene Expression Regulation ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,RNA Polymerase II ,Thiolester Hydrolases - Abstract
MYC is an oncogenic transcription factor that binds globally to active promoters and promotes transcriptional elongation by RNA polymerase II (RNAPII) 1,2 . Deregulated expression of the paralogous protein MYCN drives the development of neuronal and neuroendocrine tumours and is often associated with a particularly poor prognosis 3 . Here we show that, similar to MYC, activation of MYCN in human neuroblastoma cells induces escape of RNAPII from promoters. If the release of RNAPII from transcriptional pause sites (pause release) fails, MYCN recruits BRCA1 to promoter-proximal regions. Recruitment of BRCA1 prevents MYCN-dependent accumulation of stalled RNAPII and enhances transcriptional activation by MYCN. Mechanistically, BRCA1 stabilizes mRNA decapping complexes and enables MYCN to suppress R-loop formation in promoter-proximal regions. Recruitment of BRCA1 requires the ubiquitin-specific protease USP11, which binds specifically to MYCN when MYCN is dephosphorylated at Thr58. USP11, BRCA1 and MYCN stabilize each other on chromatin, preventing proteasomal turnover of MYCN. Because BRCA1 is highly expressed in neuronal progenitor cells during early development 4 and MYC is less efficient than MYCN in recruiting BRCA1, our findings indicate that a cell-lineage-specific stress response enables MYCN-driven tumours to cope with deregulated RNAPII function.
- Published
- 2019
18. A MYC–GCN2–eIF2α negative feedback loop limits protein synthesis to prevent MYC-dependent apoptosis in colorectal cancer
- Author
-
Martin Eilers, Elmar Wolf, Katja Maurus, Douglas Strathdee, John R. P. Knight, Apoorva Baluapuri, Florian Erhard, Friedrich Wilhelm Uthe, Andreas Rosenwald, Rene Jackstadt, Niels Matthes, Stefanie Schmidt, Madelon Paauwe, Sarah Denk, Carsten P. Ade, Catriona A. Ford, Almut Schulze, Sheila Bryson, Armin Wiegering, Owen J. Sansom, Christina Schülein-Völk, Christoph-Thomas Germer, Georgios Vlachogiannis, Nicola Valeri, Markus E. Diefenbacher, Christoph Otto, Werner Schmitz, Fiona Clare Warrander, and Susanne Walz
- Subjects
Male ,Colon ,Adenomatous Polyposis Coli Protein ,Eukaryotic Initiation Factor-2 ,eIF2α ,translation ,Apoptosis ,colorectal cancer ,MYC ,Protein Serine-Threonine Kinases ,Article ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Movement ,Cell Line, Tumor ,Protein biosynthesis ,Integrated stress response ,Animals ,Humans ,030304 developmental biology ,Cell Proliferation ,Regulation of gene expression ,Feedback, Physiological ,0303 health sciences ,Chemistry ,Kinase ,Translation (biology) ,Cell Biology ,HCT116 Cells ,Protein kinase R ,Survival Analysis ,Xenograft Model Antitumor Assays ,Cell biology ,APC ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Eukaryotic Initiation Factor-2B ,HEK293 Cells ,030220 oncology & carcinogenesis ,Protein Biosynthesis ,Phosphorylation ,Female ,eIF2B5 ,Signal transduction ,Colorectal Neoplasms ,Signal Transduction - Abstract
Tumours depend on altered rates of protein synthesis for growth and survival, which suggests that mechanisms controlling mRNA translation may be exploitable for therapy. Here, we show that loss of APC, which occurs almost universally in colorectal tumours, strongly enhances the dependence on the translation initiation factor eIF2B5. Depletion of eIF2B5 induces an integrated stress response and enhances translation of MYC via an internal ribosomal entry site. This perturbs cellular amino acid and nucleotide pools, strains energy resources and causes MYC-dependent apoptosis. eIF2B5 limits MYC expression and prevents apoptosis in APC-deficient murine and patient-derived organoids and in APC-deficient murine intestinal epithelia in vivo. Conversely, the high MYC levels present in APC-deficient cells induce phosphorylation of eIF2α via the kinases GCN2 and PKR. Pharmacological inhibition of GCN2 phenocopies eIF2B5 depletion and has therapeutic efficacy in tumour organoids, which demonstrates that a negative MYC–eIF2α feedback loop constitutes a targetable vulnerability of colorectal tumours.
- Published
- 2019
- Full Text
- View/download PDF
19. Author Correction: Reprogramming of host glutamine metabolism during Chlamydia trachomatis infection and its key role in peptidoglycan synthesis
- Author
-
Julian Fink, Francesca R Dejure, Thomas Rudel, Martin Eilers, Sudha Janaki-Raman, Maximilian Schmalhofer, Rajeeve Sivadasan, Karthika Rajeeve, Elmar Wolf, Almut Schulze, Apoorva Baluapuri, Claudia Huber, Jürgen Seibel, Werner Schmitz, Wolfgang Eisenreich, Nadine Vollmuth, and Thomas F. Wulff
- Subjects
Microbiology (medical) ,Chlamydia trachomatis infection ,Host (biology) ,Immunology ,Glutamine metabolism ,Peptidoglycan synthesis ,Cell Biology ,Biology ,Applied Microbiology and Biotechnology ,Microbiology ,Metabolomics ,Genetics ,Reprogramming - Published
- 2021
- Full Text
- View/download PDF
20. The Interaction of Myc with Miz1 Defines Medulloblastoma Subgroup Identity
- Author
-
David Finkelstein, Elmar Wolf, Yong Ha Youn, BaoHan T. Vo, Martin Eilers, Jerold E. Rehg, Susanne Walz, Martine F. Roussel, Brian L. Murphy, Anneli Gebhardt, Daisuke Kawauchi, and Young-Goo Han
- Subjects
0301 basic medicine ,Cancer Research ,animal structures ,Ubiquitin-Protein Ligases ,Article ,Proto-Oncogene Proteins c-myc ,Transcriptome ,Mice ,03 medical and health sciences ,Ciliogenesis ,medicine ,Animals ,Hedgehog Proteins ,Sonic hedgehog ,Nuclear protein ,Cerebellar Neoplasms ,neoplasms ,Neurons ,Medulloblastoma ,Regulation of gene expression ,biology ,Nuclear Proteins ,Cell Biology ,medicine.disease ,Protein Inhibitors of Activated STAT ,Chromatin ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,biology.protein ,Cancer research ,Signal Transduction - Abstract
Four distinct subgroups of cerebellar medulloblastomas (MBs) differ in their histopathology, molecular profiles, and prognosis. c-Myc (Myc) or MycN overexpression in granule neuron progenitors (GNPs) induces Group 3 (G3) or Sonic Hedgehog (SHH) MBs, respectively. Differences in Myc and MycN transcriptional profiles depend, in part, on their interaction with Miz1, which binds strongly to Myc but not MycN, to target sites on chromatin. Myc suppresses ciliogenesis and “reprograms” the transcriptome of SHH-dependent GNPs through Miz1-dependent gene repression to maintain “stemness”. Genetic disruption of the Myc/Miz1 interaction inhibited G3 MB development. Target genes of Myc/Miz1 are repressed in human G3 MBs, but not in other subgroups. Therefore, the Myc/Miz1 interaction is a defining hallmark of G3 MB development.
- Published
- 2016
- Full Text
- View/download PDF
21. Ubiquitin-Dependent Turnover of MYC Antagonizes MYC/PAF1C Complex Accumulation to Drive Transcriptional Elongation
- Author
-
Martin Eilers, Laura A. Jaenicke, Wenshan Xu, Anne Carstensen, Elmar Wolf, Björn von Eyss, Matthias Geyer, Ann Katrin Greifenberg, and Nikita Popov
- Subjects
0301 basic medicine ,BRD4 ,Time Factors ,Transcription Elongation, Genetic ,Positive Transcriptional Elongation Factor B ,Cell Cycle Proteins ,RNA polymerase II ,Transfection ,Histones ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Transactivation ,Ubiquitin ,Transcription (biology) ,Humans ,Promoter Regions, Genetic ,Transcription factor ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Binding Sites ,biology ,Tumor Suppressor Proteins ,Ubiquitination ,Nuclear Proteins ,Acetylation ,Cell Biology ,Chromatin Assembly and Disassembly ,Phosphoproteins ,Molecular biology ,HEK293 Cells ,030104 developmental biology ,Histone ,Multiprotein Complexes ,Mutation ,Proteolysis ,biology.protein ,RNA Interference ,RNA Polymerase II ,HeLa Cells ,Transcription Factors - Abstract
MYC is an unstable protein, and its turnover is controlled by the ubiquitin system. Ubiquitination enhances MYC-dependent transactivation, but the underlying mechanism remains unresolved. Here we show that MYC proteasomal turnover is dispensable for loading of RNA polymerase II (RNAPII). In contrast, MYC turnover is essential for recruitment of TRRAP, histone acetylation, and binding of BRD4 and P-TEFb to target promoters, leading to phosphorylation of RNAPII and transcriptional elongation. In the absence of histone acetylation and P-TEFb recruitment, MYC associates with the PAF1 complex (PAF1C) through a conserved domain in the MYC amino terminus ("MYC box I"). Depletion of the PAF1C subunit CDC73 enhances expression of MYC target genes, suggesting that the MYC/PAF1C complex can inhibit transcription. Because several ubiquitin ligases bind to MYC via the same domain ("MYC box II") that interacts with TRRAP, we propose that degradation of MYC limits the accumulation of MYC/PAF1C complexes during transcriptional activation.
- Published
- 2016
- Full Text
- View/download PDF
22. Localized Inhibition of Protein Phosphatase 1 by NUAK1 Promotes Spliceosome Activity and Reveals a MYC-Sensitive Feedback Control of Transcription
- Author
-
Dominik Mumberg, Guido Mastrobuoni, Apoorva Baluapuri, Bernhard Kuster, Stefan Kempa, Carsten P. Ade, Raphael Silveira Vidal, Amit Kumar, Elmar Wolf, Susanne Walz, Reinhard Lührmann, Martin Eilers, Christina Schülein-Völk, Cyrille Girard, Lars Wortmann, Yun-Chien Chang, Giacomo Cossa, Isabelle Roeschert, and Florian Prinz
- Subjects
Cancer Research ,Transcription, Genetic ,Feedback control ,Cell ,NUAK1 ,MYC ,Mice ,0302 clinical medicine ,Transcription (biology) ,Protein Phosphatase 1 ,Phosphorylation ,Nuclear protein ,0303 health sciences ,PP1 ,Chromatin ,ddc ,Cell biology ,medicine.anatomical_structure ,Spliceosome ,ARK5 ,RNA splicing ,RNA Polymerase II ,Technology Platforms ,RNA Splicing ,Protein subunit ,Phosphatase ,Biology ,Article ,Feedback ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,medicine ,PNUTS ,Animals ,Humans ,Molecular Biology ,030304 developmental biology ,Cell Nucleus ,Correction ,Protein phosphatase 1 ,Cell Biology ,Repressor Proteins ,Gene Expression Regulation ,Cardiovascular and Metabolic Diseases ,NIH 3T3 Cells ,Spliceosomes ,Protein Kinases ,030217 neurology & neurosurgery ,HeLa Cells - Abstract
Summary Deregulated expression of MYC induces a dependence on the NUAK1 kinase, but the molecular mechanisms underlying this dependence have not been fully clarified. Here, we show that NUAK1 is a predominantly nuclear protein that associates with a network of nuclear protein phosphatase 1 (PP1) interactors and that PNUTS, a nuclear regulatory subunit of PP1, is phosphorylated by NUAK1. Both NUAK1 and PNUTS associate with the splicing machinery. Inhibition of NUAK1 abolishes chromatin association of PNUTS, reduces spliceosome activity, and suppresses nascent RNA synthesis. Activation of MYC does not bypass the requirement for NUAK1 for spliceosome activity but significantly attenuates transcription inhibition. Consequently, NUAK1 inhibition in MYC-transformed cells induces global accumulation of RNAPII both at the pause site and at the first exon-intron boundary but does not increase mRNA synthesis. We suggest that NUAK1 inhibition in the presence of deregulated MYC traps non-productive RNAPII because of the absence of correctly assembled spliceosomes., Graphical Abstract, Highlights • Nuclear NUAK1 associates with PP1 and phosphorylates its targeting subunit PNUTS • NUAK1, PP1, and PNUTS form a trimer that associates with the splicing machinery • Inhibition of NUAK1 reduces spliceosome activity and nascent RNA synthesis • When MYC is deregulated, NUAK1 inhibition traps RNAPII at the intron-exon boundary, Tumors displaying high MYC levels depend on the NUAK1 kinase. Cossa et al. show that NUAK1 binds protein phosphatase 1 (PP1) and its regulatory subunit PNUTS. They show that this complex associates with the splicing machinery and provides a feedback control of transcription that can be overridden by deregulated MYC.
- Published
- 2020
- Full Text
- View/download PDF
23. Taming of the beast: shaping Myc-dependent amplification
- Author
-
Martin Eilers, Charles Y. Lin, David Levens, and Elmar Wolf
- Subjects
Genetics ,Kruppel-Like Transcription Factors ,Euchromatin ,Cell ,Genes, myc ,Cell Biology ,Biology ,medicine.disease_cause ,Phenotype ,Article ,Cell biology ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,medicine.anatomical_structure ,Transcription (biology) ,Neoplasms ,medicine ,Animals ,Humans ,Drosophila ,Carcinogenesis ,Gene ,Transcription factor - Abstract
Myc deregulation is a hallmark oncogenic event where overexpression of the transcription factor gives rise to numerous tumorigenic phenotypes. The complex consequences of Myc deregulation have prevented clear mechanistic interpretations of its function. A synthesis of recent experimental observations offers a consensus on the direct transcriptional function of Myc: when overexpressed, Myc broadly engages the established euchromatic cis-regulatory landscape of the cell, where the factor generally amplifies transcription. The level of Myc binding at target genes and the transcriptional output are differentially modulated by additional regulators, including Miz1. Targeting Myc oncogenic activity will require an understanding of whether amplification promotes tumorigenesis and the consequences of amplification in tumors adapted to oncogenic Myc.
- Published
- 2015
- Full Text
- View/download PDF
24. Association with Aurora-A Controls N-MYC-Dependent Promoter Escape and Pause Release of RNA Polymerase II during the Cell Cycle
- Author
-
Martin Eilers, Anne Carstensen, David S. Rickman, Richard Bayliss, Apoorva Baluapuri, Colin Kwok, Jacqueline Kalb, Hans Michael Maric, Steffi Herold, Elmar Wolf, Evon Poon, Ka-Yan Mak, Julia Hofstetter, Gabriele Büchel, Olga Sumara, Eoin Leen, Susanne Walz, Isabelle Roeschert, and Louis Chesler
- Subjects
0301 basic medicine ,Transcription Elongation, Genetic ,Protein subunit ,RNA polymerase II ,Cell Cycle Proteins ,Plasma protein binding ,MYC ,Aurora-A ,General Biochemistry, Genetics and Molecular Biology ,Article ,S Phase ,03 medical and health sciences ,neuroblastoma ,Transcription Factors, TFIII ,Cell Line, Tumor ,Journal Article ,Humans ,Nuclear protein ,RAD21 ,Promoter Regions, Genetic ,lcsh:QH301-705.5 ,TFIIIC ,Aurora Kinase A ,N-MYC ,N-Myc Proto-Oncogene Protein ,biology ,Effector ,Chemistry ,Nuclear Proteins ,Promoter ,Cell cycle ,Phosphoproteins ,3. Good health ,Cell biology ,Chromatin ,DNA-Binding Proteins ,030104 developmental biology ,DNA Topoisomerases, Type II ,lcsh:Biology (General) ,biology.protein ,DNA, Intergenic ,RNA Polymerase II ,pause release ,Protein Binding - Abstract
Summary MYC proteins bind globally to active promoters and promote transcriptional elongation by RNA polymerase II (Pol II). To identify effector proteins that mediate this function, we performed mass spectrometry on N-MYC complexes in neuroblastoma cells. The analysis shows that N-MYC forms complexes with TFIIIC, TOP2A, and RAD21, a subunit of cohesin. N-MYC and TFIIIC bind to overlapping sites in thousands of Pol II promoters and intergenic regions. TFIIIC promotes association of RAD21 with N-MYC target sites and is required for N-MYC-dependent promoter escape and pause release of Pol II. Aurora-A competes with binding of TFIIIC and RAD21 to N-MYC in vitro and antagonizes association of TOP2A, TFIIIC, and RAD21 with N-MYC during S phase, blocking N-MYC-dependent release of Pol II from the promoter. Inhibition of Aurora-A in S phase restores RAD21 and TFIIIC binding to chromatin and partially restores N-MYC-dependent transcriptional elongation. We propose that complex formation with Aurora-A controls N-MYC function during the cell cycle., Graphical Abstract, Highlights • N-MYC forms complexes with TFIIIC, RAD21, and TOP2A • TFIIIC recruits RAD21 and is required for N-MYC-dependent pause release of Pol II • Aurora-A displaces TFIIIC, TOP2A, and RAD21 from N-MYC during S phase • Aurora-A inhibits pause release of Pol II during S phase, Büchel et al. demonstrate that N-MYC forms complexes with TFIIIC, TOP2A, and RAD21. Aurora-A competes with TFIIIC and RAD21 for binding to N-MYC, and Aurora-A displaces the three proteins from N-MYC during S phase. As consequence, N-MYC-dependent pause release is inhibited during S phase, preventing activation of the ATR checkpoint kinase.
- Published
- 2017
- Full Text
- View/download PDF
25. PAF1 complex component Leo1 helps recruit Drosophila Myc to promoters
- Author
-
Apoorva Baluapuri, Michael Furrer, Maria Gallant, Dirk Birkel, Jennifer M. Gerlach, Peter Gallant, and Elmar Wolf
- Subjects
0301 basic medicine ,Genetics ,Multidisciplinary ,Oncogene ,Transcription, Genetic ,Promoter ,Biology ,Cell biology ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,030104 developmental biology ,PNAS Plus ,Transcription (biology) ,Animals ,Drosophila Proteins ,Drosophila ,Sequence motif ,Promoter Regions, Genetic ,Gene ,Transcription factor ,Drosophila Protein ,Cells, Cultured ,Transcription Factors - Abstract
The Myc oncogene is a transcription factor with a powerful grip on cellular growth and proliferation. The physical interaction of Myc with the E-box DNA motif has been extensively characterized, but it is less clear whether this sequence-specific interaction is sufficient for Myc's binding to its transcriptional targets. Here we identify the PAF1 complex, and specifically its component Leo1, as a factor that helps recruit Myc to target genes. Since the PAF1 complex is typically associated with active genes, this interaction with Leo1 contributes to Myc targeting to open promoters.
- Published
- 2017
26. Dual Regulation of Fbw7 Function and Oncogenic Transformation by Usp28
- Author
-
Laura A. Jänicke, Martin Eilers, Lyudmyla Taranets, Wenshan Xu, Axel Behrens, Andreas Hellmann, Markus E. Diefenbacher, Elmar Wolf, Christina Schülein-Völk, Nikita Popov, and Jing Zhu
- Subjects
F-Box-WD Repeat-Containing Protein 7 ,Transcription, Genetic ,Protein subunit ,Ubiquitin-Protein Ligases ,Cell Cycle Proteins ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Deubiquitinating enzyme ,Substrate Specificity ,Ubiquitin ,Animals ,Humans ,ddc:610 ,lcsh:QH301-705.5 ,Alleles ,Cell Proliferation ,Mice, Knockout ,biology ,Protein Stability ,F-Box Proteins ,Embryonic stem cell ,Ubiquitin ligase ,Cell biology ,Cell Transformation, Neoplastic ,Biochemistry ,lcsh:Biology (General) ,Organ Specificity ,Knockout mouse ,Proteolysis ,biology.protein ,Biocatalysis ,Ectopic expression ,Ubiquitin Thiolesterase ,Function (biology) ,Gene Deletion ,HeLa Cells - Abstract
SummaryFbw7, the substrate recognition subunit of SCF(Fbw7) ubiquitin ligase, mediates the turnover of multiple proto-oncoproteins and promotes its own degradation. Fbw7-dependent substrate ubiquitination is antagonized by the Usp28 deubiquitinase. Here, we show that Usp28 preferentially antagonizes autocatalytic ubiquitination and stabilizes Fbw7, resulting in dose-dependent effects in Usp28 knockout mice. Monoallelic deletion of Usp28 maintains stable Fbw7 but drives Fbw7 substrate degradation. In contrast, complete knockout triggers Fbw7 degradation and leads to the accumulation of Fbw7 substrates in several tissues and embryonic fibroblasts. On the other hand, overexpression of Usp28 stabilizes both Fbw7 and its substrates. Consequently, both complete loss and ectopic expression of Usp28 promote Ras-driven oncogenic transformation. We propose that dual regulation of Fbw7 activity by Usp28 is a safeguard mechanism for maintaining physiological levels of proto-oncogenic Fbw7 substrates, which is equivalently disrupted by loss or overexpression of Usp28.
- Published
- 2014
- Full Text
- View/download PDF
27. MYC Recruits SPT5 to RNA Polymerase II to Promote Processive Transcription Elongation
- Author
-
Robert Düster, Martin Eilers, Bikash Adhikari, Shuang-Yan Wang, Ashwin Narain, Elmar Wolf, Susanne Walz, Armin Wiegering, Peter Gallant, Patrick Cramer, Pranjali Bhandare, Andreas Schlosser, Seychelle M. Vos, Julia Hofstetter, Theresa Endres, Apoorva Baluapuri, Hans Michael Maric, Nevenka Dudvarski Stankovic, Matthias Geyer, Jens T. Vanselow, Jessica Denise Schwarz, Lisa Anna Jung, and Markus Vogt
- Subjects
Transcription, Genetic ,Protein subunit ,RNA polymerase II ,MYC ,Article ,elongation rate ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,directionality ,0302 clinical medicine ,Transcription (biology) ,Cell Line, Tumor ,Neoplasms ,Humans ,Histone Chaperones ,Promoter Regions, Genetic ,Molecular Biology ,Gene ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,biology ,Nuclear Proteins ,Promoter ,Cell Biology ,Processivity ,processivity ,DSIF ,Cyclin-Dependent Kinases ,SUPT5H ,Cell biology ,SPT6 ,Elongation factor ,tumorigenesis ,biology.protein ,Transcriptional Elongation Factors ,transcription ,Cyclin-Dependent Kinase-Activating Kinase ,030217 neurology & neurosurgery ,SPT5 - Abstract
Summary The MYC oncoprotein binds to promoter-proximal regions of virtually all transcribed genes and enhances RNA polymerase II (Pol II) function, but its precise mode of action is poorly understood. Using mass spectrometry of both MYC and Pol II complexes, we show here that MYC controls the assembly of Pol II with a small set of transcription elongation factors that includes SPT5, a subunit of the elongation factor DSIF. MYC directly binds SPT5, recruits SPT5 to promoters, and enables the CDK7-dependent transfer of SPT5 onto Pol II. Consistent with known functions of SPT5, MYC is required for fast and processive transcription elongation. Intriguingly, the high levels of MYC that are expressed in tumors sequester SPT5 into non-functional complexes, thereby decreasing the expression of growth-suppressive genes. Altogether, these results argue that MYC controls the productive assembly of processive Pol II elongation complexes and provide insight into how oncogenic levels of MYC permit uncontrolled cellular growth., Graphical Abstract, Highlights • MYC enhances productive transcription by defining the protein composition of Pol II • MYC directly binds SPT5 and hands it over to Pol II in a CDK7-dependent manner • Transfer of SPT5 increases speed and processivity of Pol II • MYC’s effects on Pol II function shape its tumor-specific gene expression profile, Baluapuri et al. report that MYC governs transition of Pol II into a transcriptionally engaged form by loading the SPT5 protein onto it. This increases Pol II processivity and productive transcription. The effect of MYC on Pol II can regulate expression of those genes, which help tumors to grow.
- Published
- 2019
- Full Text
- View/download PDF
28. Different promoter affinities account for specificity in MYC-dependent gene regulation
- Author
-
Uwe Benary, Jana Wolf, Francesca Lorenzin, Elmar Wolf, Martin Eilers, Björn von Eyss, Lisa Anna Jung, Caroline Kisker, Apoorva Baluapuri, and Susanne Walz
- Subjects
0301 basic medicine ,Cancer Research ,Transcription, Genetic ,WDR5 ,Mouse ,Gene Expression ,MYC ,MIZ1 ,Gene expression ,Biology (General) ,Promoter Regions, Genetic ,Cancer Biology ,Regulation of gene expression ,General Neuroscience ,mathematical modeling ,food and beverages ,General Medicine ,ChIP-sequencing ,Medicine ,Insight ,Protein Binding ,Research Article ,Human ,Chromatin Immunoprecipitation ,QH301-705.5 ,promoter affinity ,Science ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Humans ,ddc:610 ,Gene ,General Immunology and Microbiology ,Sequence Analysis, RNA ,Gene Expression Profiling ,fungi ,Epithelial Cells ,Promoter ,DNA ,Sequence Analysis, DNA ,Cell Biology ,Models, Theoretical ,Molecular biology ,Gene expression profiling ,030104 developmental biology ,Gene Expression Regulation ,Chromatin immunoprecipitation ,Transcription Factors - Abstract
Enhanced expression of the MYC transcription factor is observed in the majority of tumors. Two seemingly conflicting models have been proposed for its function: one proposes that MYC enhances expression of all genes, while the other model suggests gene-specific regulation. Here, we have explored the hypothesis that specific gene expression profiles arise since promoters differ in affinity for MYC and high-affinity promoters are fully occupied by physiological levels of MYC. We determined cellular MYC levels and used RNA- and ChIP-sequencing to correlate promoter occupancy with gene expression at different concentrations of MYC. Mathematical modeling showed that binding affinities for interactions of MYC with DNA and with core promoter-bound factors, such as WDR5, are sufficient to explain promoter occupancies observed in vivo. Importantly, promoter affinity stratifies different biological processes that are regulated by MYC, explaining why tumor-specific MYC levels induce specific gene expression programs and alter defined biological properties of cells. DOI: http://dx.doi.org/10.7554/eLife.15161.001, eLife digest Genes with the potential to cause tumors and cancer are commonly called oncogenes. One example of an oncogene encodes for a protein called MYC and many tumors contain high levels of this protein. MYC is a transcription factor and studies of aggressive tumors suggested that, like most other transcription factors, MYC binds to and regulates the activity of a small number of genes in tumors. However, other studies went on to show that MYC actually binds to thousands of genes and somehow only regulates a subset of them during tumor development. Lorenzin et al. set out to understand how this process works by generating human cells in which the concentration of MYC protein could be altered. In the experiments, the concentration was varied from normal healthy levels to the high levels found in aggressive tumors. The amount of MYC bound to genes and the extent to which it activated the genes inside these cells was also measured. Lorenzin et al. found that increasing MYC levels from normal to tumor-specific levels did not affect MYC binding at genes where the transcription factor was already strongly bound in normal cells. Rather, MYC binding increased only at genes that were weakly bound in normal cells. Consistent with this observation, only genes at which MYC was weakly bound in normal cells were activated by increasing MYC levels. This observation suggests that increasing the concentration of MYC protein from normal to tumor-specific levels “fills up” previously empty binding sites around these genes with the transcription factor. Lorenzin et al. also used mathematical modeling to understand how the concentrations of MYC in normal and tumor cells might explain how MYC behaves in cells. Together, the results imply that the MYC transcription factor regulates distinct sets of genes in normal and tumor cells according to how much MYC is present. Further studies may show that the altered regulation of a tumor-specific set of genes is important for tumor development and could use this new information to identify new targets for treating MYC-driven tumors. DOI: http://dx.doi.org/10.7554/eLife.15161.002
- Published
- 2016
29. Semiquantitative Proteomic Analysis of the Human Spliceosome via a Novel Two-Dimensional Gel Electrophoresis Method
- Author
-
Sergey Bessonov, Cindy L. Will, Elmar Wolf, Dmitry E. Agafonov, Reinhard Lührmann, Henning Urlaub, Jochen Deckert, and Peter Odenwälder
- Subjects
Proteomics ,Spliceosomal complex ,Spliceosome ,Two-dimensional gel electrophoresis ,Proteins ,Articles ,Cell Biology ,Biology ,Molecular biology ,Fluorescence ,Staining ,Electrophoresis ,Biochemistry ,RNA splicing ,Spliceosomes ,Humans ,Electrophoresis, Gel, Two-Dimensional ,Molecular Biology ,HeLa Cells - Abstract
More than 200 proteins associate with human spliceosomes, but little is known about their relative abundances in a given spliceosomal complex. Here we describe a novel two-dimensional (2D) electrophoresis method that allows separation of high-molecular-mass proteins without in-gel precipitation and thus without loss of protein. Using this system coupled with mass spectrometry, we identified 171 proteins altogether on 2D maps of stage-specific spliceosomal complexes. By staining with a fluorescent dye with a wide linear intensity range, we could quantitate and categorize proteins as present in high, moderate, or low abundance. Affinity-purified human B, B(act), and C complexes contained 69, 63, and 72 highly/moderately abundant proteins, respectively. The recruitment and release of spliceosomal proteins were followed based on their abundances in A, B, B(act), and C spliceosomal complexes. Staining with a phospho-specific dye revealed that approximately one-third of the proteins detected in human spliceosomal complexes by 2D gel analyses are phosphorylated. The 2D gel electrophoresis system described here allows for the first time an objective view of the relative abundances of proteins present in a particular spliceosomal complex and also sheds additional light on the spliceosome's compositional dynamics and the phosphorylation status of spliceosomal proteins at specific stages of splicing.
- Published
- 2011
- Full Text
- View/download PDF
30. The ubiquitin ligase Huwe1 regulates the maintenance and lymphoid commitment of hematopoietic stem cells
- Author
-
Charalampos Lazaris, Francesco Boccalatte, Kelly Moran-Crusio, Elmar Wolf, Beatriz Aranda-Orgilles, Iannis Aifantis, Xiaofeng Yu, Clarisse Kayembe, Anna Lasorella, Bryan W. King, and Jingjing Wang
- Subjects
0301 basic medicine ,Transcription, Genetic ,Ubiquitin-Protein Ligases ,Immunology ,Genes, myc ,Mice, Transgenic ,Biology ,Article ,Cell Line ,Blood cell ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Mice ,Stress, Physiological ,medicine ,Immunology and Allergy ,Animals ,Cluster Analysis ,Cell Lineage ,Lymphopoiesis ,Lymphocytes ,Cell Self Renewal ,Progenitor ,Mice, Knockout ,Protein Stability ,Gene Expression Profiling ,Tumor Suppressor Proteins ,Cell Cycle ,Cell Differentiation ,Hematopoietic Stem Cells ,Cell biology ,Ubiquitin ligase ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,biology.protein ,Bone marrow ,Stem cell - Abstract
Hematopoietic stem cells (HSCs) are dormant in the bone marrow and can be activated in response to diverse stresses to replenish all blood cell types. We identified the ubiquitin ligase Huwe1 as a crucial regulator of HSC function via its post-translational control of the oncoprotein N-myc (encoded by Mycn). We found Huwe1 to be essential for HSC self-renewal, quiescence and lymphoid-fate specification in mice. Through the use of a fluorescent fusion allele (MycnM), we observed that N-myc expression was restricted to the most immature, multipotent stem and progenitor populations. N-myc expression was upregulated in response to stress or following loss of Huwe1, which led to increased proliferation and stem-cell exhaustion. Mycn depletion reversed most of these phenotypes in vivo, which suggested that the attenuation of N-myc by Huwe1 is essential for reestablishing homeostasis following stress.
- Published
- 2015
31. LARP4B is an AU-rich sequence associated factor that promotes mRNA accumulation and translation
- Author
-
Jens T. Vanselow, Stefan Juranek, Yasuhiro Murakawa, Katrin Schäffler, Utz Fischer, Andreas Schlosser, Elmar Wolf, Maritta Küspert, and Markus Landthaler
- Subjects
Genetics ,AU-rich element ,AU Rich Elements ,Messenger RNA ,RNA ,Articles ,Biology ,Ribosome ,Autoantigens ,Polymerase Chain Reaction ,Cell biology ,Transcriptome ,HEK293 Cells ,Ribonucleoproteins ,Protein Biosynthesis ,Gene expression ,P-bodies ,Protein biosynthesis ,Humans ,RNA, Messenger ,Molecular Biology - Abstract
mRNAs are key molecules in gene expression and subject to diverse regulatory events. Regulation is accomplished by distinct sets of trans-acting factors that interact with mRNAs and form defined mRNA–protein complexes (mRNPs). The resulting “mRNP code” determines the fate of any given mRNA and thus controlling gene expression at the post-transcriptional level. The La-related protein 4B (LARP4B) belongs to an evolutionarily conserved family of RNA-binding proteins characterized by the presence of a La-module implicated in direct RNA binding. Biochemical experiments have shown previously direct interactions of LARP4B with factors of the translation machinery. This finding along with the observation of an association with actively translating ribosomes suggested that LARP4B is a factor contributing to the mRNP code. To gain insight into the function of LARP4B in vivo we tested its mRNA association at the transcriptome level and its impact on the proteome. PAR-CLIP analyses allowed us to identify the in vivo RNA targets of LARP4B. We show that LARP4B binds to a distinct set of cellular mRNAs by contacting their 3′ UTRs. Biocomputational analysis combined with in vitro binding assays identified the LARP4B-binding motif on mRNA targets. The reduction of cellular LARP4B levels leads to a marked destabilization of its mRNA targets and consequently their reduced translation. Our data identify LARP4B as a component of the mRNP code that influences the expression of its mRNA targets by affecting their stability.
- Published
- 2015
32. Miz1 is required to maintain autophagic flux
- Author
-
Anneli Gebhardt, Nicole Wagner, Georg Krohne, Martin Eilers, Esther Asan, Martine F. Roussel, Susanne Walz, Elmar Wolf, Christoph Renninger, Björn von Eyss, and Daisuke Kawauchi
- Subjects
Sequestosome-1 Protein ,Transcription, Genetic ,Ubiquitin-Protein Ligases ,General Physics and Astronomy ,Ribosome biogenesis ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Mice ,Purkinje Cells ,Cerebellum ,Transcriptional regulation ,Autophagy ,Animals ,Nuclear protein ,Promoter Regions, Genetic ,Transport Vesicles ,Transcription factor ,Heat-Shock Proteins ,Adaptor Proteins, Signal Transducing ,Zinc finger ,Mice, Knockout ,Multidisciplinary ,Binding Sites ,Integrases ,Ubiquitination ,Nuclear Proteins ,General Chemistry ,Sequence Analysis, DNA ,Protein Inhibitors of Activated STAT ,Cell biology ,Protein Structure, Tertiary ,Vesicular transport protein ,Gene Expression Regulation ,Female ,Ribosomes ,Transcription Factor TFIIH ,Protein Binding ,Signal Transduction ,Transcription Factors - Abstract
Miz1 is a zinc finger protein that regulates the expression of cell cycle inhibitors as part of a complex with Myc. Cell cycle-independent functions of Miz1 are poorly understood. Here we use a Nestin-Cre transgene to delete an essential domain of Miz1 in the central nervous system (Miz1(ΔPOZNes)). Miz1(ΔPOZNes) mice display cerebellar neurodegeneration characterized by the progressive loss of Purkinje cells. Chromatin immunoprecipitation sequencing and biochemical analyses show that Miz1 activates transcription upon binding to a non-palindromic sequence present in core promoters. Target genes of Miz1 encode regulators of autophagy and proteins involved in vesicular transport that are required for autophagy. Miz1(ΔPOZ) neuronal progenitors and fibroblasts show reduced autophagic flux. Consistently, polyubiquitinated proteins and p62/Sqtm1 accumulate in the cerebella of Miz1(ΔPOZNes) mice, characteristic features of defective autophagy. Our data suggest that Miz1 may link cell growth and ribosome biogenesis to the transcriptional regulation of vesicular transport and autophagy.
- Published
- 2013
33. Miz1 Is a Critical Repressor of cdkn1a during Skin Tumorigenesis
- Author
-
Jan Hönnemann, Adrián Sanz-Moreno, Elmar Wolf, Martin Eilers, and Hans-Peter Elsässer
- Subjects
Cell cycle checkpoint ,Skin Neoplasms ,Mouse ,Cellular differentiation ,medicine.disease_cause ,Biochemistry ,Mice ,Molecular cell biology ,Basic Cancer Research ,Genetics ,Multidisciplinary ,Kinase ,Immunochemistry ,Nuclear Proteins ,Cell Differentiation ,Animal Models ,Protein Inhibitors of Activated STAT ,Cell biology ,Cell Transformation, Neoplastic ,Oncology ,Medicine ,Tetradecanoylphorbol Acetate ,Oncology Agents ,Research Article ,Cyclin-Dependent Kinase Inhibitor p21 ,Histology ,Science ,9,10-Dimethyl-1,2-benzanthracene ,Ubiquitin-Protein Ligases ,DNA transcription ,Repressor ,Mice, Transgenic ,Biology ,Cell Growth ,Molecular Genetics ,Model Organisms ,DNA-binding proteins ,medicine ,Animals ,Gene Regulation ,ddc:610 ,Psychological repression ,Transcription factor ,Cell Proliferation ,Cyclin-Dependent Kinase Inhibitor p15 ,Cell growth ,Proteins ,Regulatory proteins ,Neoplasms, Experimental ,Protein Structure, Tertiary ,Gene expression ,Gene Function ,Carcinogenesis ,Animal Genetics - Abstract
The transcription factor Miz1 forms repressive DNA-binding complexes with the Myc, Gfi-1 and Bcl-6 oncoproteins. Known target genes of these complexes encode the cyclin-dependent kinase inhibitors (CKIs) cdkn2b (p15\(^{Ink4}\)), cdkn1a (p21\(^{Cip1}\)), and cdkn1c (p57\(^{Kip2}\)). Whether Miz1-mediated repression is important for control of cell proliferation in vivo and for tumor formation is unknown. Here we show that deletion of the Miz1 POZ domain, which is critical for Miz1 function, restrains the development of skin tumors in a model of chemically-induced, Ras-dependent tumorigenesis. While the stem cell compartment appears unaffected, interfollicular keratinocytes lacking functional Miz1 exhibit a reduced proliferation and an accelerated differentiation of the epidermis in response to the tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA). Tumorigenesis, proliferation and normal differentiation are restored in animals lacking cdkn1a, but not in those lacking cdkn2b. Our data demonstrate that Miz1-mediated attenuation of cell cycle arrest pathways via repression of cdkn1a has a critical role during tumorigenesis in the skin.
- Published
- 2012
34. 3D cryo-EM structure of an active step I spliceosome and localization of its catalytic core
- Author
-
Michael Grote, Reinhard Lührmann, Holger Stark, Sergey Bessonov, Elmar Wolf, Berthold Kastner, Monika M. Golas, and Bjoern Sander
- Subjects
Genetics ,Models, Molecular ,Spliceosome ,Cryo-electron microscopy ,Stereochemistry ,Cryoelectron Microscopy ,Intron ,Cell Biology ,Biology ,Exon ,Immunolabeling ,Catalytic Domain ,RNA splicing ,Biocatalysis ,Spliceosomes ,Humans ,snRNP ,Molecular Biology ,Ribonucleoprotein - Abstract
The spliceosome excises introns from pre-mRNA in a two-step splicing reaction. So far, the three-dimensional (3D) structure of a spliceosome with preserved catalytic activity has remained elusive. Here, we determined the 3D structure of the human, catalytically active step I spliceosome (C complex) by cryo-electron microscopy (cryo-EM) in vitrified ice. Via immunolabeling we mapped the position of the 5' exon. The C complex contains an unusually salt-stable ribonucleoprotein (RNP) core that harbors its catalytic center. We determined the 3D structure of this RNP core and also that of a post-step II particle, the 35S U5 snRNP, which contains most of the C complex core proteins. As C complex domains could be recognized in these structures, their position in the C complex could be determined, thereby allowing the region harboring the spliceosome's catalytic core to be localized.
- Published
- 2010
- Full Text
- View/download PDF
35. Molecular architecture of the human Prp19/CDC5L complex
- Author
-
Ira Lemm, Michael Grote, Henning Urlaub, Dmitry E. Agafonov, Wolfgang Fischle, Elmar Wolf, Adrian Schomburg, Cindy L. Will, and Reinhard Lührmann
- Subjects
Spliceosome ,Stereochemistry ,Saccharomyces cerevisiae ,Cell Cycle Proteins ,RNA-binding protein ,Plasma protein binding ,Blotting, Far-Western ,Models, Biological ,Chromatography, Affinity ,Protein structure ,Minor spliceosome ,Humans ,Immunoprecipitation ,Protein Structure, Quaternary ,Molecular Biology ,biology ,Protein Stability ,C-terminus ,RNA-Binding Proteins ,Articles ,Cell Biology ,biology.organism_classification ,Molecular biology ,Protein Structure, Tertiary ,Cross-Linking Reagents ,Multiprotein Complexes ,Protein Biosynthesis ,RNA splicing ,Salts ,Protein Processing, Post-Translational ,HeLa Cells ,Protein Binding - Abstract
Protein complexes containing Prp19 play a central role during catalytic activation of the spliceosome, and Prp19 and its related proteins are major components of the spliceosome's catalytic core RNP. To learn more about the spatial organization of the human Prp19 (hPrp19)/CDC5L complex, which is comprised of hPrp19, CDC5L, PRL1, AD002, SPF27, CTNNBL1, and HSP73, we purified native hPrp19/CDC5L complexes from HeLa cells stably expressing FLAG-tagged AD002 or SPF27. Stoichiometric analyses indicated that, like Saccharomyces cerevisiae NTC (nineteen complex), the human Prp19/CDC5L complex contains four copies of hPrp19. Salt treatment identified a stable core comprised of CDC5L, hPrp19, PRL1, and SPF27. Protein-protein interaction studies revealed that SPF27 directly interacts with each component of the hPrp19/CDC5L complex core and also elucidated several additional, previously unknown interactions between hPrp19/CDC5L complex components. Limited proteolysis of the hPrp19/CDC5L complex revealed a protease-resistant complex comprised of SPF27, the C terminus of CDC5L, and the N termini of PRL1 and hPrp19. Under the electron microscope, purified hPrp19/CDC5L complexes exhibit an elongated, asymmetric shape with a maximum dimension of approximately 20 nm. Our findings not only elucidate the molecular organization of the hPrp19/CDC5L complex but also provide insights into potential protein-protein interactions at the core of the catalytically active spliceosome.
- Published
- 2010
36. Conservation of the Protein Composition and Electron Microscopy Structure of Drosophila melanogaster and Human Spliceosomal Complexes▿ †
- Author
-
Cindy L. Will, Berthold Kastner, Reinhard Lührmann, Nadine Herold, Elmar Wolf, and Henning Urlaub
- Subjects
Cell Extracts ,Spliceosome ,RNA Splicing ,Fushi Tarazu Transcription Factors ,Proteomics ,Chromatography, Affinity ,Conserved sequence ,Substrate Specificity ,RNA Precursors ,Animals ,Drosophila Proteins ,Humans ,Molecular Biology ,Conserved Sequence ,Genetics ,Cell Nucleus ,biology ,Intron ,Nuclear Proteins ,Cell Biology ,Articles ,Exons ,Aptamers, Nucleotide ,biology.organism_classification ,Introns ,Cell biology ,DNA-Binding Proteins ,Kinetics ,Microscopy, Electron ,Drosophila melanogaster ,Ribonucleoproteins ,RNA splicing ,Spliceosomes ,Exon junction complex ,Drosophila Protein ,HeLa Cells - Abstract
Comprehensive proteomics analyses of spliceosomal complexes are currently limited to those in humans, and thus, it is unclear to what extent the spliceosome's highly complex composition and compositional dynamics are conserved among metazoans. Here we affinity purified Drosophila melanogaster spliceosomal B and C complexes formed in Kc cell nuclear extract. Mass spectrometry revealed that their composition is highly similar to that of human B and C complexes. Nonetheless, a number of Drosophila-specific proteins were identified, suggesting that there may be novel factors contributing specifically to splicing in flies. Protein recruitment and release events during the B-to-C transition were also very similar in both organisms. Electron microscopy of Drosophila B complexes revealed a high degree of structural similarity with human B complexes, indicating that higher-order interactions are also largely conserved. A comparison of Drosophila spliceosomes formed on a short versus long intron revealed only small differences in protein composition but, nonetheless, clear structural differences under the electron microscope. Finally, the characterization of affinity-purified Drosophila mRNPs indicated that exon junction complex proteins are recruited in a splicing-dependent manner during C complex formation. These studies provide insights into the evolutionarily conserved composition and structure of the metazoan spliceosome, as well as its compositional dynamics during catalytic activation.
- Published
- 2008
37. Localization of Prp8, Brr2, Snu114 and U4/U6 proteins in the yeast tri-snRNP by electron microscopy
- Author
-
Holger Stark, Reinhard Lührmann, Elif Karagöz, Elmar Wolf, Monika M. Golas, Bjoern Sander, Berthold Kastner, Irina Häcker, and Patrizia Fabrizio
- Subjects
Spliceosome ,Saccharomyces cerevisiae Proteins ,Macromolecular Substances ,Protein Conformation ,Ribonucleoprotein, U4-U6 Small Nuclear ,Saccharomyces cerevisiae ,GTPase ,Biology ,environment and public health ,03 medical and health sciences ,0302 clinical medicine ,Structural Biology ,snRNP ,Molecular Biology ,Ribonucleoprotein, U5 Small Nuclear ,030304 developmental biology ,Ribonucleoprotein ,Genetics ,0303 health sciences ,biology.organism_classification ,Ribonucleoproteins, Small Nuclear ,RNA Helicase A ,Cell biology ,Enzyme Activation ,Microscopy, Electron ,RNA splicing ,Spliceosomes ,Nucleic Acid Conformation ,030217 neurology & neurosurgery ,Small nuclear RNA ,RNA Helicases - Abstract
The tri-snRNP is the largest preassembled unit of the spliceosome, and its components are key to the splicing reaction. The overall structure and conformations of the yeast tri-snRNP are now analyzed by EM, and the general positions of some of its major protein components mapped. The U4/U6-U5 tri–small nuclear ribonucleoprotein (snRNP) is a major, evolutionarily highly conserved spliceosome subunit. Unwinding of its U4/U6 snRNA duplex is a central event of spliceosome activation that requires several components of the U5 portion of the tri-snRNP, including the RNA helicase Brr2, Prp8 and the GTPase Snu114. Here we report the EM projection structure of the Saccharomyces cerevisiae tri-snRNP. It shows a modular organization comprising three extruding domains that contact one another in its central portion. We have visualized genetically tagged tri-snRNP proteins by EM and show here that U4/U6 snRNP forms a domain termed the arm. Conversely, a separate head domain adjacent to the arm harbors Brr2, whereas Prp8 and the GTPase Snu114 are located centrally. The head and arm adopt variable relative positions. This molecular organization and dynamics suggest possible scenarios for structural events during catalytic activation.
- Published
- 2008
38. GraFix: sample preparation for single-particle electron cryomicroscopy
- Author
-
Henning Urlaub, Holger Stark, Klaus Hartmuth, Franz Herzog, Berthold Kastner, Reinhard Lührmann, Prakash Dube, Jochen Deckert, Dietmar Poerschke, Florian Hauer, Jan-Michael Peters, Daniel Boehringer, Hannes Uchtenhagen, Monika M. Golas, Bjoern Sander, Elmar Wolf, and Niels Fischer
- Subjects
Tissue Fixation ,Materials science ,Chromatography ,Cryo-electron microscopy ,Cryoelectron Microscopy ,Cell Biology ,Gradient centrifugation ,Image Enhancement ,Biochemistry ,Specimen Handling ,Sample quality ,Reagent ,Sample preparation ,Molecular Biology ,Biotechnology ,Macromolecule - Abstract
We developed a method, named GraFix, that considerably improves sample quality for structure determination by single-particle electron cryomicroscopy (cryo-EM). GraFix uses a glycerol gradient centrifugation step in which the complexes are centrifuged into an increasing concentration of a chemical fixation reagent to prevent aggregation and to stabilize individual macromolecules. The method can be used to prepare samples for negative-stain, cryo-negative-stain and, particularly, unstained cryo-EM.
- Published
- 2008
39. Target gene-independent functions of MYC oncoproteins
- Author
-
Martin Eilers, Elmar Wolf, and Apoorva Baluapuri
- Subjects
DNA Replication ,Transcription, Genetic ,Carcinogenesis ,Biology ,medicine.disease_cause ,Article ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,0302 clinical medicine ,Transcription (biology) ,Neoplasms ,medicine ,Humans ,Molecular Biology ,Transcription factor ,Gene ,Cell Proliferation ,030304 developmental biology ,Oncogene Proteins ,N-Myc Proto-Oncogene Protein ,0303 health sciences ,General transcription factor ,Cell Cycle ,DNA replication ,RNA ,Cell Biology ,Cell biology ,030220 oncology & carcinogenesis ,Target gene ,Transcription Factors - Abstract
Oncoproteins of the MYC family are major drivers of human tumorigenesis. Since a large body of evidence indicates that MYC proteins are transcription factors, studying their function has focused on the biology of their target genes. Detailed studies of MYC-dependent changes in RNA levels have provided contrasting models of the oncogenic activity of MYC proteins through either enhancing or repressing the expression of specific target genes, or as global amplifiers of transcription. In this Review, we first summarize the biochemistry of MYC proteins and what is known (or is unclear) about the MYC target genes. We then discuss recent progress in defining the interactomes of MYC and MYCN and how this information affects central concepts of MYC biology, focusing on mechanisms by which MYC proteins modulate transcription. MYC proteins promote transcription termination upon stalling of RNA polymerase II, and we propose that this mechanism enhances the stress resilience of basal transcription. Furthermore, MYC proteins coordinate transcription elongation with DNA replication and cell cycle progression. Finally, we argue that the mechanism by which MYC proteins regulate the transcription machinery is likely to promote tumorigenesis independently of global or relative changes in the expression of their target genes. The MYC oncoproteins are transcription factors, but the molecular mechanism of their oncogenic activity is unclear. MYC proteins promote transcription termination in stress conditions, which is proposed to increase cellular resilience to stress and to promote tumorigenesis independently of changes in the expression of their target genes.
- Full Text
- View/download PDF
40. Design, Synthesis, and Evaluation of WD-Repeat-Containing Protein 5 (WDR5) Degraders
- Author
-
Andreas Krämer, Elmar Wolf, Frank Löhr, Dalia Barsyte-Lovejoy, Martin Eilers, Mathias Diebold, Magdalena M. Szewczyk, Christoph A. Sotriffer, Cheryl H. Arrowsmith, Lena-Marie Berger, Stephanie Heinzlmeir, Jakob Gebel, Bernhard Kuster, Volker Dötsch, Stefan Knapp, Bikash Adhikari, Anja Dölle, Janik Weckesser, and Nicola Berner
- Subjects
Male ,Scaffold protein ,Dihydropyridines ,Antineoplastic Agents ,Ligands ,01 natural sciences ,Structure-Activity Relationship ,03 medical and health sciences ,Drug Discovery ,Humans ,WDR5 ,Gene ,Transcription factor ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,Dose-Response Relationship, Drug ,Molecular Structure ,biology ,Chemistry ,WD Repeat-Containing Protein 5 ,Biphenyl Compounds ,Intracellular Signaling Peptides and Proteins ,0104 chemical sciences ,Cell biology ,010404 medicinal & biomolecular chemistry ,Histone ,Drug Design ,Histone methyltransferase ,biology.protein ,Molecular Medicine ,Female ,Linker - Abstract
Histone H3K4 methylation serves as a post-translational hallmark of actively transcribed genes and is introduced by histone methyltransferase (HMT) and its regulatory scaffolding proteins. One of these is the WD-repeat-containing protein 5 (WDR5) that has also been associated with controlling long noncoding RNAs and transcription factors including MYC. The wide influence of dysfunctional HMT complexes and the typically upregulated MYC levels in diverse tumor types suggested WDR5 as an attractive drug target. Indeed, protein-protein interface inhibitors for two protein interaction interfaces on WDR5 have been developed. While such compounds only inhibit a subset of WDR5 interactions, chemically induced proteasomal degradation of WDR5 might represent an elegant way to target all oncogenic functions. This study presents the design, synthesis, and evaluation of two diverse WDR5 degrader series based on two WIN site binding scaffolds and shows that linker nature and length strongly influence degradation efficacy.
- Full Text
- View/download PDF
41. Ubiquitylation of MYC couples transcription elongation with double-strand break repair at active promoters
- Author
-
Seychelle M. Vos, Jan B. Heidelberger, Peter Gallant, Petra Beli, Nikita Popov, Elmar Wolf, Carsten P. Ade, Johannes Zuber, Matthias Muhar, Valentina Andrioletti, Ursula Eilers, Theresa Endres, Daniel Solvie, Apoorva Baluapuri, Martin Eilers, and Patrick Cramer
- Subjects
Transcription Elongation, Genetic ,DNA Repair ,Ubiquitin-Protein Ligases ,RNA polymerase II ,Article ,Histones ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Cell Line, Tumor ,Histone H2B ,Humans ,DNA Breaks, Double-Stranded ,Promoter Regions, Genetic ,Molecular Biology ,Polymerase ,030304 developmental biology ,0303 health sciences ,biology ,Tumor Suppressor Proteins ,Ubiquitination ,Promoter ,Cell Biology ,Double Strand Break Repair ,Chromatin ,Ubiquitin ligase ,Cell biology ,biology.protein ,ATPases Associated with Diverse Cellular Activities ,RNA Polymerase II ,030217 neurology & neurosurgery - Abstract
The MYC oncoprotein globally affects the function of RNA polymerase II (RNAPII). The ability of MYC to promote transcription elongation depends on its ubiquitylation. Here, we show that MYC and PAF1c (polymerase II-associated factor 1 complex) interact directly and mutually enhance each other's association with active promoters. PAF1c is rapidly transferred from MYC onto RNAPII. This transfer is driven by the HUWE1 ubiquitin ligase and is required for MYC-dependent transcription elongation. MYC and HUWE1 promote histone H2B ubiquitylation, which alters chromatin structure both for transcription elongation and double-strand break repair. Consistently, MYC suppresses double-strand break accumulation in active genes in a strictly PAF1c-dependent manner. Depletion of PAF1c causes transcription-dependent accumulation of double-strand breaks, despite widespread repair-associated DNA synthesis. Our data show that the transfer of PAF1c from MYC onto RNAPII efficiently couples transcription elongation with double-strand break repair to maintain the genomic integrity of MYC-driven tumor cells.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.