58 results on '"Michael Howell"'
Search Results
2. Identifying SARS-CoV-2 antiviral compounds by screening for small molecule inhibitors of nsp14/nsp10 exoribonuclease
- Author
-
Berta Canal, Allison W. McClure, Joseph F. Curran, Mary Wu, Rachel Ulferts, Florian Weissmann, Jingkun Zeng, Agustina P. Bertolin, Jennifer C. Milligan, Souradeep Basu, Lucy S. Drury, Tom Deegan, Ryo Fujisawa, Emma L. Roberts, Clovis Basier, Karim Labib, Rupert Beale, Michael Howell, and John F.X Diffley
- Subjects
viruses ,Drug Evaluation, Preclinical ,Viral Nonstructural Proteins ,Biochemistry ,Antiviral Agents ,Fluorescence ,Small Molecule Libraries ,03 medical and health sciences ,Biochemical Techniques & Resources ,Virology ,Chlorocebus aethiops ,Animals ,Viral Regulatory and Accessory Proteins ,Molecular Biology ,Vero Cells ,Research Articles ,030304 developmental biology ,Enzyme Assays ,0303 health sciences ,SARS-CoV-2 ,030302 biochemistry & molecular biology ,Aurintricarboxylic Acid ,Reproducibility of Results ,Cell Biology ,High-Throughput Screening Assays ,Patulin ,Exoribonucleases - Abstract
SummarySARS-CoV-2 is a coronavirus that emerged in 2019 and rapidly spread across the world causing a deadly pandemic with tremendous social and economic costs. Healthcare systems worldwide are under great pressure, and there is urgent need for effective antiviral treatments. The only currently approved antiviral treatment for COVID-19 is remdesivir, an inhibitor of viral genome replication. SARS-CoV-2 proliferation relies on the enzymatic activities of the non-structural proteins (nsp), which makes them interesting targets for the development of new antiviral treatments. With the aim to identify novel SARS-CoV-2 antivirals, we have purified the exoribonuclease/methyltransferase (nsp14) and its cofactor (nsp10) and developed biochemical assays compatible with high-throughput approaches to screen for exoribonuclease inhibitors. We have screened a library of over 5000 commercial compounds and identified patulin and aurintricarboxylic acid (ATA) as inhibitors of nsp14 exoribonuclease in vitro. We found that patulin and ATA inhibit replication of SARS-CoV-2 in a VERO E6 cell-culture model. These two new antiviral compounds will be valuable tools for further coronavirus research as well as potentially contributing to new therapeutic opportunities for COVID-19.
- Published
- 2021
3. Identifying SARS-CoV-2 antiviral compounds by screening for small molecule inhibitors of Nsp14 RNA cap methyltransferase
- Author
-
Kang Wei Tan, Emma L. Roberts, Tiffany Mak, Lucy S. Drury, Souradeep Basu, Berta Canal, Michael Howell, Joseph F. Curran, Florian Weissmann, Karim Labib, Tom D Deegan, John F.X. Diffley, Allison W McClure, Ryo Fujisawa, Mary Wu, Victoria H. Cowling, Rachel Ulferts, Clovis Basier, Rupert Beale, Theresa U. Zeisner, and Chew Theng Lim
- Subjects
Methyltransferase ,Indoles ,coronavirus ,Drug Evaluation, Preclinical ,Viral Nonstructural Proteins ,medicine.disease_cause ,Biochemistry ,Substrate Specificity ,Phenothiazines ,Chlorocebus aethiops ,Fluorescence Resonance Energy Transfer ,Coronaviridae ,Viral Regulatory and Accessory Proteins ,Research Articles ,Coronavirus ,0303 health sciences ,Alanine ,Translation (biology) ,Small molecule ,covid-19 ,Indenes ,mRNA cap ,RNA Caps ,Indazoles ,Viral protein ,Biology ,Chlorobenzenes ,Antiviral Agents ,Small Molecule Libraries ,03 medical and health sciences ,Biochemical Techniques & Resources ,Trifluperidol ,Virology ,Nitriles ,medicine ,Animals ,Molecular Biology ,Vero Cells ,030304 developmental biology ,Enzyme Assays ,030306 microbiology ,SARS-CoV-2 ,RNA ,Reproducibility of Results ,Cell Biology ,Methyltransferases ,biology.organism_classification ,Adenosine Monophosphate ,High-Throughput Screening Assays ,Viral replication ,Purines ,Exoribonucleases ,methyltransferase - Abstract
The COVID-19 pandemic has presented itself as one of the most critical public health challenges of the century, with SARS-CoV-2 being the third member of the Coronaviridae family to cause a fatal disease in humans. There is currently only one antiviral compound, remdesivir, that can be used for the treatment of COVID-19. To identify additional potential therapeutics, we investigated the enzymatic proteins encoded in the SARS-CoV-2 genome. In this study, we focussed on the viral RNA cap methyltransferases, which play key roles in enabling viral protein translation and facilitating viral escape from the immune system. We expressed and purified both the guanine-N7 methyltransferase nsp14, and the nsp16 2′-O-methyltransferase with its activating cofactor, nsp10. We performed an in vitro high-throughput screen for inhibitors of nsp14 using a custom compound library of over 5000 pharmaceutical compounds that have previously been characterised in either clinical or basic research. We identified four compounds as potential inhibitors of nsp14, all of which also showed antiviral capacity in a cell-based model of SARS-CoV-2 infection. Three of the four compounds also exhibited synergistic effects on viral replication with remdesivir.
- Published
- 2021
4. Identifying SARS-CoV-2 antiviral compounds by screening for small molecule inhibitors of nsp15 endoribonuclease
- Author
-
Berta Canal, Tom D Deegan, Karim Labib, Agustina P Bertolin, Rupert Beale, John F.X. Diffley, Florian Weissmann, Michael Howell, Ryo Fujisawa, Mary Wu, Lucy S. Drury, Rachel Ulferts, Allison W McClure, and Jingkun Zeng
- Subjects
0301 basic medicine ,medicine.drug_class ,Endoribonuclease activity ,Endoribonuclease ,coronavirus ,Drug Evaluation, Preclinical ,Biology ,In Vitro Techniques ,Viral Nonstructural Proteins ,medicine.disease_cause ,Biochemistry ,Antiviral Agents ,endonuclease ,Fluorescence ,Chemical library ,Small Molecule Libraries ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immune system ,Biochemical Techniques & Resources ,Allosteric Regulation ,Exoribonuclease ,Virology ,Chlorocebus aethiops ,Endoribonucleases ,medicine ,Animals ,Molecular Biology ,Vero Cells ,Research Articles ,Coronavirus ,Enzyme Assays ,SARS-CoV-2 ,COVID-19 ,Reproducibility of Results ,Cell Biology ,High-Throughput Screening Assays ,Solutions ,Kinetics ,030104 developmental biology ,chemistry ,nsp15 ,030220 oncology & carcinogenesis ,Vero cell ,Antiviral drug ,Viral genome replication ,Naphthoquinones - Abstract
SummarySARS-CoV-2 is responsible for COVID-19, a human disease that has caused over 2 million deaths, stretched health systems to near-breaking point and endangered the economies of countries and families around the world. Antiviral treatments to combat COVID-19 are currently lacking. Remdesivir, the only antiviral drug approved for the treatment of COVID-19, can affect disease severity, but better treatments are needed. SARS-CoV-2 encodes 16 non-structural proteins (nsp) that possess different enzymatic activities with important roles in viral genome replication, transcription and host immune evasion. One key aspect of host immune evasion is performed by the uridine-directed endoribonuclease activity of nsp15. Here we describe the expression and purification of nsp15 recombinant protein. We have developed biochemical assays to follow its activity, and we have found evidence for allosteric behaviour. We screened a custom chemical library of over 5000 compounds to identify nsp15 endoribonuclease inhibitors, and we identified and validated NSC95397 as an inhibitor of nsp15 endoribonuclease in vitro. Although NSC95397 did not inhibit SARS-CoV-2 growth in VERO E6 cells, further studies will be required to determine the effect of nsp15 inhibition on host immune evasion.
- Published
- 2021
5. Identifying SARS-CoV-2 antiviral compounds by screening for small molecule inhibitors of Nsp3 papain-like protease
- Author
-
Chew Theng Lim, Kang Wei Tan, Mary Wu, Rachel Ulferts, Lee A. Armstrong, Eiko Ozono, Lucy S. Drury, Jennifer C. Milligan, Theresa U. Zeisner, Jingkun Zeng, Florian Weissmann, Berta Canal, Ganka Bineva-Todd, Michael Howell, Nicola O'Reilly, Rupert Beale, Yogesh Kulathu, Karim Labib, and John F.X. Diffley
- Subjects
0301 basic medicine ,viruses ,coronavirus ,Drug Evaluation, Preclinical ,Coronavirus Papain-Like Proteases ,Naphthalenes ,Virus Replication ,Biochemistry ,Antiviral Agents ,Small Molecule Libraries ,03 medical and health sciences ,Inhibitory Concentration 50 ,0302 clinical medicine ,Biochemical Techniques & Resources ,Virology ,Flavins ,ubiquitin ,Chlorocebus aethiops ,Fluorescence Resonance Energy Transfer ,Animals ,Furans ,Molecular Biology ,Vero Cells ,Research Articles ,Enzyme Assays ,Alanine ,Aniline Compounds ,SARS-CoV-2 ,Quinones ,COVID-19 ,Reproducibility of Results ,protease ,Drug Synergism ,Cell Biology ,Phenanthrenes ,Adenosine Monophosphate ,High-Throughput Screening Assays ,030104 developmental biology ,030220 oncology & carcinogenesis ,Benzamides - Abstract
The COVID-19 pandemic has emerged as the biggest life-threatening disease of this century. Whilst vaccination should provide a long-term solution, this is pitted against the constant threat of mutations in the virus rendering the current vaccines less effective. Consequently, small molecule antiviral agents would be extremely useful to complement the vaccination program. The causative agent of COVID-19 is a novel coronavirus, SARS-CoV-2, which encodes at least nine enzymatic activities that all have drug targeting potential. The papain-like protease (PLpro) contained in the nsp3 protein generates viral non-structural proteins from a polyprotein precursor, and cleaves ubiquitin and ISG protein conjugates. Here we describe the expression and purification of PLpro. We developed a protease assay that was used to screen a custom compound library from which we identified dihydrotanshinone I and Ro 08-2750 as compounds that inhibit PLpro in protease and isopeptidase assays and also inhibit viral replication in cell culture-based assays.
- Published
- 2021
6. Microbial communities form rich extracellular metabolomes that foster metabolic interactions and promote drug tolerance
- Author
-
Jason S. L. Yu, Clara Correia-Melo, Francisco Zorrilla, Lucia Herrera-Dominguez, Mary Y. Wu, Johannes Hartl, Kate Campbell, Sonja Blasche, Marco Kreidl, Anna-Sophia Egger, Christoph B. Messner, Vadim Demichev, Anja Freiwald, Michael Mülleder, Michael Howell, Judith Berman, Kiran R. Patil, Mohammad Tauqeer Alam, Markus Ralser, Yu, Jason SL [0000-0001-5203-3603], Correia-Melo, Clara [0000-0001-6062-1472], Herrera-Dominguez, Lucia [0000-0001-8276-2241], Wu, Mary Y [0000-0002-2074-6171], Hartl, Johannes [0000-0001-8470-5355], Egger, Anna-Sophia [0000-0002-5204-7121], Howell, Michael [0000-0003-0912-0079], Berman, Judith [0000-0002-8577-0084], Alam, Mohammad Tauqeer [0000-0002-6872-0691], Ralser, Markus [0000-0001-9535-7413], Apollo - University of Cambridge Repository, and Apollo-University Of Cambridge Repository
- Subjects
Microbiology (medical) ,631/326/2565/855 ,RM ,Immunology ,Applied Microbiology and Biotechnology ,Microbiology ,82/80 ,13/44 ,14/34 ,Ecology,Evolution & Ethology ,38/47 ,Genetics ,14/35 ,Computational & Systems Biology ,Chemical Biology & High Throughput ,82/58 ,Microbiota ,article ,Cell Biology ,Drug Tolerance ,631/326/22 ,QP ,96/63 ,QR ,13/31 ,Metabolism ,Metabolome ,bacteria ,Microbial Interactions ,Synthetic Biology ,Metabolic Networks and Pathways - Abstract
Funder: United Arab Emirates University (UAEU); doi: https://doi.org/10.13039/501100006013, Microbial communities are composed of cells of varying metabolic capacity, and regularly include auxotrophs that lack essential metabolic pathways. Through analysis of auxotrophs for amino acid biosynthesis pathways in microbiome data derived from >12,000 natural microbial communities obtained as part of the Earth Microbiome Project (EMP), and study of auxotrophic-prototrophic interactions in self-establishing metabolically cooperating yeast communities (SeMeCos), we reveal a metabolically imprinted mechanism that links the presence of auxotrophs to an increase in metabolic interactions and gains in antimicrobial drug tolerance. As a consequence of the metabolic adaptations necessary to uptake specific metabolites, auxotrophs obtain altered metabolic flux distributions, export more metabolites and, in this way, enrich community environments in metabolites. Moreover, increased efflux activities reduce intracellular drug concentrations, allowing cells to grow in the presence of drug levels above minimal inhibitory concentrations. For example, we show that the antifungal action of azoles is greatly diminished in yeast cells that uptake metabolites from a metabolically enriched environment. Our results hence provide a mechanism that explains why cells are more robust to drug exposure when they interact metabolically.
- Published
- 2022
- Full Text
- View/download PDF
7. A network of transcription factors governs the dynamics of NODAL/Activin transcriptional responses
- Author
-
Davide M. Coda, Harshil Patel, Ilaria Gori, Tessa E. Gaarenstroom, Ok-Ryul Song, Michael Howell, and Caroline S. Hill
- Subjects
Model organisms ,smad ,animal structures ,Nodal Protein ,pathways ,Gene Expression ,Smad2 Protein ,Signalling & Oncogenes ,Transforming Growth Factor beta ,atac-seq ,expression ,Humans ,primitive streak ,Computational & Systems Biology ,Chemical Biology & High Throughput ,tgf-beta ,zic3 ,Genome Integrity & Repair ,Gene Expression Regulation, Developmental ,Membrane Proteins ,activin ,differentiation ,Cell Biology ,Tumour Biology ,proteins ,Chromatin ,foxi3 ,Activins ,wnt ,nodal ,transcription ,Genetics & Genomics ,Developmental Biology ,Transcription Factors - Abstract
SMAD2, an effector of the NODAL/Activin signalling pathway, regulates developmental processes by sensing distinct chromatin states and interacting with different transcriptional partners. However, the network of factors that controls SMAD2 chromatin binding and shapes its transcriptional programme over time is poorly characterised. Here, we combine ATAC-seq with computational footprinting to identify temporal changes in chromatin accessibility and transcription factor activity upon NODAL/Activin signalling. We show that SMAD2 binding induces chromatin opening genome wide. We discover footprints for FOXI3, FOXO3 and ZIC3 at the SMAD2-bound enhancers of the early response genes, Pmepa1 and Wnt3, respectively, and demonstrate their functionality. Finally, we determine a mechanism by which NODAL/Activin signalling induces delayed gene expression, by uncovering a self-enabling transcriptional cascade whereby activated SMADs, together with ZIC3, induce the expression of Wnt3. The resultant activated WNT pathway then acts together with the NODAL/Activin pathway to regulate expression of delayed target genes in prolonged NODAL/Activin signalling conditions. This article has an associated First Person interview with the first author of the paper.
- Published
- 2022
8. SARS-CoV-2 S2-targeted vaccination elicits broadly neutralizing antibodies
- Author
-
Kevin W. Ng, Nikhil Faulkner, Katja Finsterbusch, Mary Wu, Ruth Harvey, Saira Hussain, Maria Greco, Yafei Liu, Svend Kjaer, Charles Swanton, Sonia Gandhi, Rupert Beale, Steve J. Gamblin, Peter Cherepanov, John McCauley, Rodney Daniels, Michael Howell, Hisashi Arase, Andreas Wack, David L.V. Bauer, and George Kassiotis
- Subjects
Model organisms ,COVID-19 Vaccines ,Immunology ,Infectious Disease ,Antibodies, Viral ,Biochemistry & Proteomics ,Coronavirus OC43, Human ,Mice ,Ecology,Evolution & Ethology ,Animals ,Humans ,Computational & Systems Biology ,Chemical Biology & High Throughput ,Human Biology & Physiology ,SARS-CoV-2 ,FOS: Clinical medicine ,Stem Cells ,Vaccination ,Genome Integrity & Repair ,Neurosciences ,COVID-19 ,General Medicine ,Cell Biology ,Tumour Biology ,Antibodies, Neutralizing ,Metabolism ,Spike Glycoprotein, Coronavirus ,Cell Cycle & Chromosomes ,Genetics & Genomics ,Broadly Neutralizing Antibodies ,Structural Biology & Biophysics - Abstract
Several variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have emerged during the current coronavirus disease 2019 (COVID-19) pandemic. Although antibody cross-reactivity with the spike glycoproteins (S) of diverse coronaviruses, including endemic common cold coronaviruses (HCoVs), has been documented, it remains unclear whether such antibody responses, typically targeting the conserved S2 subunit, contribute to protection when induced by infection or through vaccination. Using a mouse model, we found that prior HCoV-OC43 S–targeted immunity primes neutralizing antibody responses to otherwise subimmunogenic SARS-CoV-2 S exposure and promotes S2-targeting antibody responses. Moreover, vaccination with SARS-CoV-2 S2 elicited antibodies in mice that neutralized diverse animal and human alphacoronaviruses and betacoronaviruses in vitro and provided a degree of protection against SARS-CoV-2 challenge in vivo. Last, in mice with a history of SARS-CoV-2 Wuhan–based S vaccination, further S2 vaccination induced broader neutralizing antibody response than booster Wuhan S vaccination, suggesting that it may prevent repertoire focusing caused by repeated homologous vaccination. These data establish the protective value of an S2-targeting vaccine and support the notion that S2 vaccination may better prepare the immune system to respond to the changing nature of the S1 subunit in SARS-CoV-2 variants of concern, as well as to future coronavirus zoonoses.
- Published
- 2022
- Full Text
- View/download PDF
9. Functional immune responses against SARS-CoV-2 variants of concern after fourth COVID-19 vaccine dose or infection in patients with blood cancer
- Author
-
Annika Fendler, Scott T.C. Shepherd, Lewis Au, Mary Wu, Ruth Harvey, Katalin A. Wilkinson, Andreas M. Schmitt, Zayd Tippu, Benjamin Shum, Sheima Farag, Aljosja Rogiers, Eleanor Carlyle, Kim Edmonds, Lyra Del Rosario, Karla Lingard, Mary Mangwende, Lucy Holt, Hamid Ahmod, Justine Korteweg, Tara Foley, Taja Barber, Andrea Emslie-Henry, Niamh Caulfield-Lynch, Fiona Byrne, Daqi Deng, Svend Kjaer, Ok-Ryul Song, Christophe J. Queval, Caitlin Kavanagh, Emma C. Wall, Edward J. Carr, Simon Caidan, Mike Gavrielides, James I. MacRae, Gavin Kelly, Kema Peat, Denise Kelly, Aida Murra, Kayleigh Kelly, Molly O’Flaherty, Robyn L. Shea, Gail Gardner, Darren Murray, Sanjay Popat, Nadia Yousaf, Shaman Jhanji, Kate Tatham, David Cunningham, Nicholas Van As, Kate Young, Andrew J.S. Furness, Lisa Pickering, Rupert Beale, Charles Swanton, Sonia Gandhi, Steve Gamblin, David L.V. Bauer, George Kassiotis, Michael Howell, Emma Nicholson, Susanna Walker, Robert J. Wilkinson, James Larkin, and Samra Turajlic
- Subjects
Model organisms ,COVID-19 Vaccines ,Immunology ,T cells ,Infectious Disease ,variants of concern ,Antibodies, Viral ,Biochemistry & Proteomics ,General Biochemistry, Genetics and Molecular Biology ,Signalling & Oncogenes ,Ecology,Evolution & Ethology ,Neoplasms ,Humans ,neutralizing antibodies ,BNT162 Vaccine ,Computational & Systems Biology ,Chemical Biology & High Throughput ,Human Biology & Physiology ,SARS-CoV-2 ,blood cancer ,FOS: Clinical medicine ,Stem Cells ,Clinical Studies as Topic ,Genome Integrity & Repair ,Immunity ,Neurosciences ,COVID-19 ,Cell Biology ,Tumour Biology ,Antibodies, Neutralizing ,Metabolism ,Cell Cycle & Chromosomes ,Genetics & Genomics ,Developmental Biology ,Structural Biology & Biophysics - Abstract
Patients with blood cancer continue to have a greater risk of inadequate immune responses following three COVID-19 vaccine doses and risk of severe COVID-19 disease. In the context of the CAPTURE study (NCT03226886), we report immune responses in 80 patients with blood cancer who received a fourth dose of BNT162b2. We measured neutralizing antibody titers (NAbTs) using a live virus microneutralization assay against wild-type (WT), Delta, and Omicron BA.1 and BA.2 and T cell responses against WT and Omicron BA.1 using an activation-induced marker (AIM) assay. The proportion of patients with detectable NAb titers and T cell responses after the fourth vaccine dose increased compared with that after the third vaccine dose. Patients who received B cell-depleting therapies within the 12 months before vaccination have the greatest risk of not having detectable NAbT. In addition, we report immune responses in 57 patients with breakthrough infections after vaccination.
- Published
- 2022
- Full Text
- View/download PDF
10. USP25 promotes pathological HIF-1-driven metabolic reprogramming and is a potential therapeutic target in pancreatic cancer
- Author
-
Jessica K. Nelson, May Zaw Thin, Theodore Evan, Steven Howell, Mary Wu, Bruna Almeida, Nathalie Legrave, Duco S. Koenis, Gabriela Koifman, Yoichiro Sugimoto, Miriam Llorian Sopena, James MacRae, Emma Nye, Michael Howell, Ambrosius P. Snijders, Andreas Prachalias, Yoh Zen, Debashis Sarker, and Axel Behrens
- Subjects
Model organisms ,endocrine system diseases ,Immunology ,General Physics and Astronomy ,Gene Expression ,Infectious Disease ,Biochemistry & Proteomics ,General Biochemistry, Genetics and Molecular Biology ,Signalling & Oncogenes ,Mice ,Cell Line, Tumor ,Tumor Microenvironment ,Animals ,Humans ,Computational & Systems Biology ,Chemical Biology & High Throughput ,Human Biology & Physiology ,Multidisciplinary ,Stem Cells ,FOS: Clinical medicine ,Genome Integrity & Repair ,General Chemistry ,Cell Biology ,Tumour Biology ,digestive system diseases ,Pancreatic Neoplasms ,Metabolism ,Cardiovascular and Metabolic Diseases ,Genetics & Genomics ,Glycolysis ,Ubiquitin Thiolesterase ,Developmental Biology ,Carcinoma, Pancreatic Ductal - Abstract
Deubiquitylating enzymes (DUBs) play an essential role in targeted protein degradation and represent an emerging therapeutic paradigm in cancer. However, their therapeutic potential in pancreatic ductal adenocarcinoma (PDAC) has not been explored. Here, we develop a DUB discovery pipeline, combining activity-based proteomics with a loss-of-function genetic screen in patient-derived PDAC organoids and murine genetic models. This approach identifies USP25 as a master regulator of PDAC growth and maintenance. Genetic and pharmacological USP25 inhibition results in potent growth impairment in PDAC organoids, while normal pancreatic organoids are insensitive, and causes dramatic regression of patient-derived xenografts. Mechanistically, USP25 deubiquitinates and stabilizes the HIF-1α transcription factor. PDAC is characterized by a severely hypoxic microenvironment, and USP25 depletion abrogates HIF-1α transcriptional activity and impairs glycolysis, inducing PDAC cell death in the tumor hypoxic core. Thus, the USP25/HIF-1α axis is an essential mechanism of metabolic reprogramming and survival in PDAC, which can be therapeutically exploited.
- Published
- 2021
11. Identifying SARS-CoV-2 Antiviral Compounds by Screening for Small Molecule Inhibitors of Nsp12/7/8 RNA-dependent RNA Polymerase
- Author
-
Florian Weissmann, Berta Canal, Rachel Ulferts, Mary Wu, Michael Howell, John F.X. Diffley, Lucy S. Drury, Jennifer C. Milligan, Rupert Beale, Jingkun Zeng, Agustina P Bertolin, and Viktor Posse
- Subjects
0301 basic medicine ,viruses ,Drug Evaluation, Preclinical ,coronavirus ,Druggability ,Viral Nonstructural Proteins ,medicine.disease_cause ,RNA-dependent RNA polymerase ,Biochemistry ,Benzoates ,Chemical library ,chemistry.chemical_compound ,0302 clinical medicine ,RNA polymerase ,Chlorocebus aethiops ,Fluorescence Resonance Energy Transfer ,Research Articles ,Coronavirus ,chemistry.chemical_classification ,Coronavirus RNA-Dependent RNA Polymerase ,biology ,Small molecule ,Suramin ,Antiviral Agents ,Small Molecule Libraries ,03 medical and health sciences ,Biochemical Techniques & Resources ,Virology ,medicine ,Animals ,Molecular Biology ,Vero Cells ,Enzyme Assays ,SARS-CoV-2 ,Reproducibility of Results ,COVID-19 ,RNA virus ,Cell Biology ,Bridged Bicyclo Compounds, Heterocyclic ,biology.organism_classification ,High-Throughput Screening Assays ,nsp12 ,030104 developmental biology ,Enzyme ,Viral replication ,chemistry ,Holoenzymes ,030217 neurology & neurosurgery - Abstract
SummaryThe coronavirus disease 2019 (COVID-19) global pandemic has turned into the largest public health and economic crisis in recent history impacting virtually all sectors of society. There is a need for effective therapeutics to battle the ongoing pandemic. Repurposing existing drugs with known pharmacological safety profiles is a fast and cost-effective approach to identify novel treatments. The COVID-19 etiologic agent is the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a single-stranded positive-sense RNA virus. Coronaviruses rely on the enzymatic activity of the replication-transcription complex (RTC) to multiply inside host cells. The RTC core catalytic component is the RNA-dependent RNA polymerase (RdRp) holoenzyme. The RdRp is one of the key druggable targets for CoVs due to its essential role in viral replication, high degree of sequence and structural conservation and the lack of homologs in human cells. Here, we have expressed, purified and biochemically characterised active SARS-CoV-2 RdRp complexes. We developed a novel fluorescence resonance energy transfer (FRET)-based strand displacement assay for monitoring SARS-CoV-2 RdRp activity suitable for a high-throughput format. As part of a larger research project to identify inhibitors for all the enzymatic activities encoded by SARS-CoV-2, we used this assay to screen a custom chemical library of over 5000 approved and investigational compounds for novel SARS-CoV-2 RdRp inhibitors. We identified 3 novel compounds (GSK-650394, C646 and BH3I-1) and confirmed suramin and suramin-like compounds as in vitro SARS-CoV-2 RdRp activity inhibitors. We also characterised the antiviral efficacy of these drugs in cell-based assays that we developed to monitor SARS-CoV-2 growth.
- Published
- 2021
12. Identifying SARS-CoV-2 Antiviral Compounds by Screening for Small Molecule Inhibitors of Nsp13 Helicase
- Author
-
John W. McCauley, Ruth Harvey, Mary Wu, Laura E. McCoy, Agustina P Bertolin, Viktor Posse, Rupert Beale, Lucy S. Drury, John F.X. Diffley, Svend Kjaer, Saira Hussain, Berta Canal, Annabel Borg, Florian Weissmann, Jingkun Zeng, Jennifer C. Milligan, Rachel Ulferts, Michael Howell, and Chloe Roustan
- Subjects
0301 basic medicine ,viruses ,Drug Evaluation, Preclinical ,coronavirus ,Viral Nonstructural Proteins ,medicine.disease_cause ,Biochemistry ,Chemical library ,chemistry.chemical_compound ,0302 clinical medicine ,Chlorocebus aethiops ,Fluorescence Resonance Energy Transfer ,Research Articles ,media_common ,Coronavirus ,0303 health sciences ,biology ,030302 biochemistry & molecular biology ,RNA Helicase A ,Small molecule ,RNA Helicases ,Drug ,RNA helicase ,media_common.quotation_subject ,Suramin ,Antiviral Agents ,Virus ,Small Molecule Libraries ,03 medical and health sciences ,Biochemical Techniques & Resources ,Virology ,High-Throughput Screening Assays ,medicine ,Animals ,Molecular Biology ,Vero Cells ,Enzyme Assays ,030304 developmental biology ,SARS-CoV-2 ,Reproducibility of Results ,COVID-19 ,Helicase ,Cell Biology ,030104 developmental biology ,Viral replication ,chemistry ,nsp13 ,Vero cell ,biology.protein ,030217 neurology & neurosurgery - Abstract
SummaryThe coronavirus disease 2019 (COVID-19) pandemic, which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a global public health challenge. While the efficacy of vaccines against emerging and future virus variants remains unclear, there is a need for therapeutics. Repurposing existing drugs represents a promising and potentially rapid opportunity to find novel antivirals against SARS-CoV-2. The virus encodes at least nine enzymatic activities that are potential drug targets. Here we have expressed, purified and developed enzymatic assays for SARS-CoV-2 nsp13 helicase, a viral replication protein that is essential for the coronavirus life cycle. We screened a custom chemical library of over 5000 previously characterised pharmaceuticals for nsp13 inhibitors using a FRET-based high-throughput screening (HTS) approach. From this, we have identified FPA-124 and several suramin-related compounds as novel inhibitors of nsp13 helicase activity in vitro. We describe the efficacy of these drugs using assays we developed to monitor SARS-CoV-2 growth in Vero E6 cells.
- Published
- 2021
13. Identifying SARS-CoV-2 Antiviral Compounds by Screening for Small Molecule Inhibitors of nsp5 Main Protease
- Author
-
Florian Weissmann, Chew Theng Lim, Clovis Basier, Christelle Soudy, Nicola O’Reilly, Karim Labib, Tom D Deegan, Ganka Bineva-Todd, Theresa U. Zeisner, Kang Wei Tan, Berta Canal, Rachel Ulferts, John F.X. Diffley, Dhira Joshi, Rupert Beale, Jennifer C. Milligan, Joseph F. Curran, George Papageorgiou, Michael Howell, Mary Wu, Ryo Fujisawa, and Hema Nagaraj
- Subjects
Azoles ,Leupeptins ,Peptidomimetic ,viruses ,medicine.medical_treatment ,Drug Evaluation, Preclinical ,coronavirus ,Isoindoles ,Viral Nonstructural Proteins ,medicine.disease_cause ,Biochemistry ,Amino Acid Chloromethyl Ketones ,Chemical library ,chemistry.chemical_compound ,0302 clinical medicine ,Organoselenium Compounds ,Chlorocebus aethiops ,nsp5 ,Fluorescence Resonance Energy Transfer ,Coronavirus 3C Proteases ,Research Articles ,Coronavirus ,0303 health sciences ,RNA-Binding Proteins ,Antiviral Agents ,Small Molecule Libraries ,03 medical and health sciences ,Biochemical Techniques & Resources ,Virology ,medicine ,Animals ,Vero Cells ,Molecular Biology ,Enzyme Assays ,030304 developmental biology ,Protease ,SARS-CoV-2 ,Leupeptin ,Reproducibility of Results ,COVID-19 ,protease ,Cell Biology ,In vitro ,High-Throughput Screening Assays ,Viral replication ,chemistry ,Vero cell ,Peptidomimetics ,030217 neurology & neurosurgery - Abstract
SummaryThe coronavirus 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), spread around the world with unprecedented health and socio-economic effects for the global population. While different vaccines are now being made available, very few antiviral drugs have been approved. The main viral protease (nsp5) of SARS-CoV-2 provides an excellent target for antivirals, due to its essential and conserved function in the viral replication cycle. We have expressed, purified and developed assays for nsp5 protease activity. We screened the nsp5 protease against a custom chemical library of over 5,000 characterised pharmaceuticals. We identified calpain inhibitor I and three different peptidyl fluoromethylketones (FMK) as inhibitors of nsp5 activityin vitro, with IC50values in the low micromolar range. By altering the sequence of our peptidomimetic FMK inhibitors to better mimic the substrate sequence of nsp5, we generated an inhibitor with a subnanomolar IC50. Calpain inhibitor I inhibited viral infection in monkey-derived Vero E6 cells, with an EC50 in the low micromolar range. The most potent and commercially available peptidyl-FMK compound inhibited viral growth in Vero E6 cells to some extent, while our custom peptidyl FMK inhibitor offered a marked antiviral improvement.
- Published
- 2021
14. Correction: WDHD1 is essential for the survival of PTEN-inactive triple-negative breast cancer
- Author
-
David C. Hancock, Charlotte Hill, Julian Downward, Paul Skipp, Marcin R. Przewloka, Rob M. Ewing, Yihua Wang, Huiquan Liu, Xianglin Yuan, Ayse Ertay, Hua Xiong, Mark J. Coldwell, Dian Liu, Michael Howell, and Ping Peng
- Subjects
Cancer Research ,Immunology ,Triple Negative Breast Neoplasms ,Cellular and Molecular Neuroscience ,Breast cancer ,Text mining ,Cell Line, Tumor ,medicine ,Humans ,PTEN ,lcsh:QH573-671 ,Triple-negative breast cancer ,biology ,lcsh:Cytology ,business.industry ,PTEN Phosphohydrolase ,Correction ,Cell Biology ,Middle Aged ,medicine.disease ,DNA-Binding Proteins ,Cancer research ,biology.protein ,Female ,business ,Cell signalling ,Signal Transduction - Abstract
Triple-negative breast cancer (TNBC) is the most aggressive type of breast cancer that lacks the oestrogen receptor, progesterone receptor and human epidermal growth factor receptor 2, making it difficult to target therapeutically. Targeting synthetic lethality is an alternative approach for cancer treatment. TNBC shows frequent loss of phosphatase and tensin homologue (PTEN) expression, which is associated with poor prognosis and treatment response. To identify PTEN synthetic lethal interactions, TCGA analysis coupled with a whole-genome siRNA screen in isogenic PTEN-negative and -positive cells were performed. Among the candidate genes essential for the survival of PTEN-inactive TNBC cells, WDHD1 (WD repeat and high-mobility group box DNA-binding protein 1) expression was increased in the low vs. high PTEN TNBC samples. It was also the top hit in the siRNA screen and its knockdown significantly inhibited cell viability in PTEN-negative cells, which was further validated in 2D and 3D cultures. Mechanistically, WDHD1 is important to mediate a high demand of protein translation in PTEN-inactive TNBC. Finally, the importance of WDHD1 in TNBC was confirmed in patient samples obtained from the TCGA and tissue microarrays with clinic-pathological information. Taken together, as an essential gene for the survival of PTEN-inactive TNBC cells, WDHD1 could be a potential biomarker or a therapeutic target for TNBC.
- Published
- 2021
15. Mechanism of Rac and Cdc42 Synchronization at the Cell Edge by ARHGAP39-Dependent Signaling and the Impact on Protrusion Dynamics
- Author
-
Christopher Welch, Céline DerMardirossian, Ritu Pathak, Violaine Delorme-Walker, Klaus M. Hahn, and Michael Howell
- Subjects
animal structures ,GTPase-activating protein ,Chemistry ,Cell migration ,macromolecular substances ,CDC42 ,Compartmentalization (psychology) ,Cell biology ,embryonic structures ,Phosphorylation ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Lamellipodium ,Filopodia - Abstract
Compartmentalization of GTPase regulators into signaling nodules dictates specific GTPase signaling pathway selection. Rac and Cdc42 are synchronized at the cell edge for effective protrusion in motile cells, but how their activity is coordinated remains elusive. Here, we report that ARHGAP39, a Rac and Cdc42 GTPase-activating protein, sequentially interacts with WAVE and mDia2 to control lamellipodia and filopodia protrusions, respectively. Mechanistically, ARHGAP39 binds WAVE, whereupon phosphorylation by Src promotes Rac inactivation and leads to Cdc42-induced filopodia formation. Using an optimized FRET biosensor, we detected active Cdc42 at the filopodia tips that controls filopodia extension. ARHGAP39 is transported to filopodia tips by Myosin-X where it binds mDia2 and inactivates Cdc42, leading to filopodia retraction. Failure in lamellipodia-to-filopodia switch by defective ARHGAP39 impairs cell invasion and metastasis. Our study reveals that compartmentalization of ARHGAP39 within Rac/Cdc42 signaling nodules orchestrates the synchronization of lamellipodia and filopodia and highlights the intricate regulation of leading edge dynamics in migrating cells.
- Published
- 2021
16. SMAD proteins: Mediators of diverse outcomes during infection
- Author
-
Lachlan Yuek Shun Lai, Nicholas Peter Gracie, Anjali Gowripalan, Liam Michael Howell, and Timothy Peter Newsome
- Subjects
Histology ,QH573-671 ,Pathogen ,Transforming Growth Factor β ,Smad Proteins ,Cell Biology ,General Medicine ,Fibrosis ,Pathology and Forensic Medicine ,Cellular Signalling ,Transforming Growth Factor beta ,Humans ,Infection ,Cytology ,Receptors, Transforming Growth Factor beta ,SMAD ,Signal Transduction - Abstract
Understanding the relationship between host and pathogen is key to combatting disease. SMAD transcription factors, which transmit TGF-β superfamily signalling, mediate an array of outcomes during embryogenesis, inflammation, cancer, and immunity. Surprisingly, these activities can sometimes be directly opposed; for example, SMAD3 has been reported as tumour suppressor by arresting cell cycle progression but conversely promotes cancer metastasis. A growing body of literature has identified SMADs as prominent targets during viral and bacterial infection for modulating host signalling. During infection, the activity of SMAD-containing transcriptional complexes can be finely tuned by pathogens to enhance infectivity and spread. SMAD signalling can be modulated at many levels, such as upstream at the ligand and receptor, or by direct interactions with SMADs. These alterations can increase pathogen dissemination, induce fibrosis, over-activate, or attenuate the host immune response. Here, we summarise the diverse mechanisms by which pathogens have evolved to sway SMAD signalling in their favour. Understanding the intricacies of host-pathogen interactions through this lens may elucidate aspects of SMAD function in cancer development, homoeostasis, and immune signalling previously overlooked. These insights are an opportunity to identify novel TGF-β or BMP-targeted therapeutics for applications to infectious disease contexts.
- Published
- 2022
17. WDHD1 is essential for the survival of PTEN-inactive triple negative breast cancer
- Author
-
Julian Downward, Ping Peng, Huiquan Liu, Paul Skipp, Rob M. Ewing, Michael Howell, Hua Xiong, Marcin R. Przewloka, Mark J. Coldwell, Dian Liu, Yihua Wang, Xianglin Yuan, David C. Hancock, Charlotte Hill, and Ayse Ertay
- Subjects
Cancer Research ,Gene knockdown ,Tissue microarray ,biology ,lcsh:Cytology ,Immunology ,Cell Biology ,Synthetic lethality ,medicine.disease ,Article ,Cellular and Molecular Neuroscience ,Breast cancer ,Progesterone receptor ,biology.protein ,medicine ,Cancer research ,PTEN ,Tensin ,lcsh:QH573-671 ,Triple-negative breast cancer ,Cell signalling - Abstract
Triple-negative breast cancer (TNBC) is the most aggressive type of breast cancer that lacks the oestrogen receptor, progesterone receptor and human epidermal growth factor receptor 2, making it difficult to target therapeutically. Targeting synthetic lethality is an alternative approach for cancer treatment. TNBC shows frequent loss of phosphatase and tensin homologue (PTEN) expression, which is associated with poor prognosis and treatment response. To identify PTEN synthetic lethal interactions, TCGA analysis coupled with a whole-genome siRNA screen in isogenic PTEN-negative and -positive cells were performed. Among the candidate genes essential for the survival of PTEN-inactive TNBC cells, WDHD1 (WD repeat and high-mobility group box DNA-binding protein 1) expression was increased in the low vs. high PTEN TNBC samples. It was also the top hit in the siRNA screen and its knockdown significantly inhibited cell viability in PTEN-negative cells, which was further validated in 2D and 3D cultures. Mechanistically, WDHD1 is important to mediate a high demand of protein translation in PTEN-inactive TNBC. Finally, the importance of WDHD1 in TNBC was confirmed in patient samples obtained from the TCGA and tissue microarrays with clinic-pathological information. Taken together, as an essential gene for the survival of PTEN-inactive TNBC cells, WDHD1 could be a potential biomarker or a therapeutic target for TNBC.
- Published
- 2020
18. Hairpin RNA-induced conformational change of a eukaryotic-specific lysyl-tRNA synthetase extension and role of adjacent anticodon-binding domain
- Author
-
Shuohui Liu, Michael Howell, Andrew B. Herr, Jennifer M. Hinerman, Sheng Liu, Karin Musier-Forsyth, Maryanne Refaei, Pearl Tsang, and Aaron Decker
- Subjects
0301 basic medicine ,Lysine-tRNA Ligase ,Models, Molecular ,Conformational change ,RNA Folding ,Magnetic Resonance Spectroscopy ,Aminoacylation ,Biochemistry ,Small hairpin RNA ,03 medical and health sciences ,chemistry.chemical_compound ,Protein Domains ,RNA, Transfer ,Humans ,Molecular Biology ,030102 biochemistry & molecular biology ,Aminoacyl tRNA synthetase ,RNA ,Cell Biology ,TRNA binding ,030104 developmental biology ,chemistry ,Transfer RNA ,Protein Structure and Folding ,Biophysics ,Binding domain - Abstract
Human lysyl-tRNA synthetase (hLysRS) is essential for aminoacylation of tRNA(Lys). Higher eukaryotic LysRSs possess an N-terminal extension (Nterm) previously shown to facilitate high-affinity tRNA binding and aminoacylation. This eukaryote-specific appended domain also plays a critical role in hLysRS nuclear localization, thus facilitating noncanonical functions of hLysRS. The structure is intrinsically disordered and therefore remains poorly characterized. Findings of previous studies are consistent with the Nterm domain undergoing a conformational transition to an ordered structure upon nucleic acid binding. In this study, we used NMR to investigate how the type of RNA, as well as the presence of the adjacent anticodon-binding domain (ACB), influences the Nterm conformation. To explore the latter, we used sortase A ligation to produce a segmentally labeled tandem-domain protein, Nterm–ACB. In the absence of RNA, Nterm remained disordered regardless of ACB attachment. Both alone and when attached to ACB, Nterm structure remained unaffected by titration with single-stranded RNAs. The central region of the Nterm domain adopted α-helical structure upon titration of Nterm and Nterm–ACB with RNA hairpins containing double-stranded regions. Nterm binding to the RNA hairpins resulted in CD spectral shifts consistent with an induced helical structure. NMR and fluorescence anisotropy revealed that Nterm binding to hairpin RNAs is weak but that the binding affinity increases significantly upon covalent attachment to ACB. We conclude that the ACB domain facilitates induced-fit conformational changes and confers high-affinity RNA hairpin binding, which may be advantageous for functional interactions of LysRS with a variety of different binding partners.
- Published
- 2020
19. Control of skeletal morphogenesis by the Hippo-YAP/TAZ pathway
- Author
-
Rebecca E. Saunders, Oriane Guillermin, Timothy J. Mohun, Alexander Howson, Fabrice Prin, Thomas Snoeks, Barry J. Thompson, Michael Howell, Bishara Marzook, Hannah Vanyai, and Stefan Boeing
- Subjects
TAZ ,Hippo pathway ,Morphogenesis ,Cell Cycle Proteins ,Cartilage morphogenesis ,Biology ,Protein Serine-Threonine Kinases ,Mouse embryo ,Chondrocyte ,Bone and Bones ,Extracellular matrix ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Chondrocytes ,medicine ,Animals ,Hippo Signaling Pathway ,Growth Plate ,Molecular Biology ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Cell Nucleus ,Mice, Knockout ,0303 health sciences ,Hippo signaling pathway ,Cartilage ,Tumor Suppressor Proteins ,Gene Expression Regulation, Developmental ,YAP-Signaling Proteins ,Cell biology ,Extracellular Matrix ,CTGF ,Cleft Palate ,Mice, Inbred C57BL ,medicine.anatomical_structure ,CYR61 ,Trans-Activators ,YAP ,030217 neurology & neurosurgery ,Developmental Biology ,Signal Transduction ,Research Article - Abstract
The Hippo-YAP/TAZ pathway is an important regulator of tissue growth, but can also control cell fate or tissue morphogenesis. Here, we investigate the function of the Hippo pathway during the development of cartilage, which forms the majority of the skeleton. Previously, YAP was proposed to inhibit skeletal size by repressing chondrocyte proliferation and differentiation. We find that, in vitro, Yap/Taz double knockout impairs murine chondrocyte proliferation, whereas constitutively nuclear nls-YAP5SA accelerates proliferation, in line with the canonical role of this pathway in most tissues. However, in vivo, cartilage-specific knockout of Yap/Taz does not prevent chondrocyte proliferation, differentiation or skeletal growth, but rather results in various skeletal deformities including cleft palate. Cartilage-specific expression of nls-YAP5SA or knockout of Lats1/2 do not increase cartilage growth, but instead lead to catastrophic malformations resembling chondrodysplasia or achondrogenesis. Physiological YAP target genes in cartilage include Ctgf, Cyr61 and several matrix remodelling enzymes. Thus, YAP/TAZ activity controls chondrocyte proliferation in vitro, possibly reflecting a regenerative response, but is dispensable for chondrocyte proliferation in vivo, and instead functions to control cartilage morphogenesis via regulation of the extracellular matrix., Summary: The primary role of Hippo-YAP/TAZ signalling in cartilage development is in control of tissue morphogenesis, rather than in control of cell proliferation or cell fate.
- Published
- 2019
20. The glutathione redox system is essential to prevent ferroptosis caused by impaired lipid metabolism in clear cell renal cell carcinoma
- Author
-
Beatrice Dankworth, Ming Jiang, Michael Howell, Dean W. Felsher, Arvin M. Gouw, Becky Saunders, Barrie Peck, Julian Downward, Mathias T. Rosenfeldt, Werner Schmitz, Heike Miess, and Almut Schulze
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,Biology ,Article ,Lipid peroxidation ,03 medical and health sciences ,chemistry.chemical_compound ,Cell Line, Tumor ,Basic Helix-Loop-Helix Transcription Factors ,Genetics ,medicine ,Humans ,Carcinoma, Renal Cell ,Molecular Biology ,Cell Proliferation ,Glutathione Peroxidase ,Cell Death ,Cell growth ,Lipid metabolism ,Glutathione ,Hypoxia-Inducible Factor 1, alpha Subunit ,Lipid Metabolism ,medicine.disease ,Kidney Neoplasms ,Cell biology ,Clear cell renal cell carcinoma ,030104 developmental biology ,chemistry ,Hypoxia-inducible factors ,Cancer cell ,Lipid Peroxidation ,Oxidation-Reduction - Abstract
Metabolic reprogramming is a prominent feature of clear cell renal cell carcinoma (ccRCC). Here we investigated metabolic dependencies in a panel of ccRCC cell lines using nutrient depletion, functional RNAi screening and inhibitor treatment. We found that ccRCC cells are highly sensitive to the depletion of glutamine or cystine, two amino acids required for glutathione (GSH) synthesis. Moreover, silencing of enzymes of the GSH biosynthesis pathway or glutathione peroxidases, which depend on GSH for the removal of cellular hydroperoxides, selectively reduced viability of ccRCC cells but did not affect the growth of non-malignant renal epithelial cells. Inhibition of GSH synthesis triggered ferroptosis, an iron-dependent form of cell death associated with enhanced lipid peroxidation. VHL is a major tumour suppressor in ccRCC and loss of VHL leads to stabilisation of hypoxia inducible factors HIF-1α and HIF-2α. Restoration of functional VHL via exogenous expression of pVHL reverted ccRCC cells to an oxidative metabolism and rendered them insensitive to the induction of ferroptosis. VHL reconstituted cells also exhibited reduced lipid storage and higher expression of genes associated with oxidiative phosphorylation and fatty acid metabolism. Importantly, inhibition of β-oxidation or mitochondrial ATP-synthesis restored ferroptosis sensitivity in VHL reconstituted cells. We also found that inhibition of GSH synthesis blocked tumour growth in a MYC-dependent mouse model of renal cancer. Together, our data suggest that reduced fatty acid metabolism due to inhibition of β-oxidation renders renal cancer cells highly dependent on the GSH/GPX pathway to prevent lipid peroxidation and ferroptotic cell death.
- Published
- 2018
21. GATA3 Mediates a Fast, Irreversible Commitment to BMP4-Driven Differentiation in Human Embryonic Stem Cells
- Author
-
Rahuman S. Malik Sheriff, Michael Howell, Borzo Gharibi, Adrienne E. Sullivan, Roy Wollman, Amelia Edwards, Ming Jiang, Alexandra Gunne-Braden, Alok Kumar Maity, Yi-Fang Wang, Robert J. Goldstone, Silvia D.M. Santos, and Philip East
- Subjects
bistability ,Human Embryonic Stem Cells ,BMP4 ,Bone Morphogenetic Protein 4 ,GATA3 Transcription Factor ,Biology ,Medical and Health Sciences ,Article ,03 medical and health sciences ,0302 clinical medicine ,GATA3 ,Genetics ,Humans ,030304 developmental biology ,Positive feedback ,0303 health sciences ,commitment ,fate decisions ,positive feedback ,Cell Differentiation ,differentiation ,Cell Biology ,Biological Sciences ,Embryonic stem cell ,Cell biology ,Bone morphogenetic protein 4 ,hESC ,embryonic structures ,Molecular Medicine ,030217 neurology & neurosurgery ,Developmental Biology ,Signal Transduction - Abstract
Summary During early development, extrinsic triggers prompt pluripotent cells to begin the process of differentiation. When and how human embryonic stem cells (hESCs) irreversibly commit to differentiation is a fundamental yet unanswered question. By combining single-cell imaging, genomic approaches, and mathematical modeling, we find that hESCs commit to exiting pluripotency unexpectedly early. We show that bone morphogenetic protein 4 (BMP4), an important differentiation trigger, induces a subset of early genes to mirror the sustained, bistable dynamics of upstream signaling. Induction of one of these genes, GATA3, drives differentiation in the absence of BMP4. Conversely, GATA3 knockout delays differentiation and prevents fast commitment to differentiation. We show that positive feedback at the level of the GATA3-BMP4 axis induces fast, irreversible commitment to differentiation. We propose that early commitment may be a feature of BMP-driven fate choices and that interlinked feedback is the molecular basis for an irreversible transition from pluripotency to differentiation., Graphical Abstract, Highlights • Irreversible commitment to BMP4-driven hESC differentiation is fast • SMAD activation is sustained, bistable, and irreversible due to positive feedback • GATA3 mirrors SMAD dynamics and mediates fast commitment to differentiation • GATA3 is an early commitment gene, Gunne-Braden et al. show that GATA3 is an early commitment gene that mediates fast commitment to BMP4-driven differentiation in human embryonic stem cells (hESCs). Interlinked feedback regulation at the level of the GATA3-BMP4 axis allows for an irreversible exit from pluripotency and an early commitment to differentiation.
- Published
- 2020
22. WISp39 binds phosphorylated Coronin 1B to regulate Arp2/3 localization and Cofilin-dependent motility
- Author
-
Michael Howell, Robert L. Margolis, Justin J Choi, Arun Fotedar, Howard Brickner, Céline DerMardirossian, Andreas Panopoulos, Daniel Miller, Rati Fotedar, Jean-Michel Saffin, and Violaine D. Delorme-Walker
- Subjects
Coronin ,Motility ,macromolecular substances ,Plasma protein binding ,Actin-Related Protein 2-3 Complex ,Article ,Tacrolimus Binding Proteins ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Cell Line, Tumor ,Phosphoprotein Phosphatases ,Humans ,HSP90 Heat-Shock Proteins ,Immunophilins ,Phosphorylation ,RNA, Small Interfering ,Research Articles ,Actin ,030304 developmental biology ,0303 health sciences ,Slingshot ,biology ,Microfilament Proteins ,Cell Biology ,Cofilin ,Cell biology ,Enzyme Activation ,HEK293 Cells ,Actin Depolymerizing Factors ,biology.protein ,RNA Interference ,030217 neurology & neurosurgery ,HeLa Cells ,Protein Binding - Abstract
WISp39 associates with Hsp90, Coronin 1B, and Slingshot phosphatase to regulate Cofilin activation and Arp2/3 complex localization at the leading edge of migrating cells., We previously identified Waf1 Cip1 stabilizing protein 39 (WISp39) as a binding partner for heat shock protein 90 (Hsp90). We now report that WISp39 has an essential function in the control of directed cell migration, which requires WISp39 interaction with Hsp90. WISp39 knockdown (KD) resulted in the loss of directional motility of mammalian cells and profound changes in cell morphology, including the loss of a single leading edge. WISp39 binds Coronin 1B, known to regulate the Arp2/3 complex and Cofilin at the leading edge. WISp39 preferentially interacts with phosphorylated Coronin 1B, allowing it to complex with Slingshot phosphatase (SSH) to dephosphorylate and activate Cofilin. WISp39 also regulates Arp2/3 complex localization at the leading edge. WISp39 KD-induced morphological changes could be rescued by overexpression of Coronin 1B together with a constitutively active Cofilin mutant. We conclude that WISp39 associates with Hsp90, Coronin 1B, and SSH to regulate Cofilin activation and Arp2/3 complex localization at the leading edge.
- Published
- 2015
23. Acetyl-CoA Synthetase 2 Promotes Acetate Utilization and Maintains Cancer Cell Growth under Metabolic Stress
- Author
-
Eric O. Aboagye, Lynn McGarry, Zachary T. Schug, Niels J. F. van den Broek, Karen Blyth, Barrie Peck, Michael J.O. Wakelam, Michael Howell, Francois Lassailly, Shaun E. Grosskurth, Gillian M. Mackay, Israt S. Alam, May Zaw Thin, Elizabeth Smethurst, Ming Jiang, Almut Schulze, Dylan T. Jones, Louise Goodwin, Eyal Gottlieb, Vinay Bulusu, Susan E. Critchlow, Bradley Spencer-Dene, Qifeng Zhang, David P. Strachan, Susan M. Mason, Emma Shanks, Adrian L. Harris, Rebecca E. Saunders, Jurre J. Kamphorst, Gabriela Kalna, Daniel James, Saverio Tardito, and Gordon Stamp
- Subjects
Cancer Research ,Gene Dosage ,Acetate-CoA Ligase ,Mice, Nude ,Biology ,Article ,Mice ,Stress, Physiological ,Cell Line, Tumor ,Neoplasms ,Lipidomics ,ACSS2 ,Gene silencing ,Animals ,Humans ,Hypoxia ,Cell Proliferation ,chemistry.chemical_classification ,Tumor microenvironment ,Cell growth ,Fatty Acids ,Fatty acid ,Cell Biology ,Acetyl—CoA synthetase ,Gene Expression Regulation, Neoplastic ,Oncology ,Biochemistry ,chemistry ,Cancer cell ,Disease Progression ,MCF-7 Cells ,Neoplasm Transplantation - Abstract
Summary A functional genomics study revealed that the activity of acetyl-CoA synthetase 2 (ACSS2) contributes to cancer cell growth under low-oxygen and lipid-depleted conditions. Comparative metabolomics and lipidomics demonstrated that acetate is used as a nutritional source by cancer cells in an ACSS2-dependent manner, and supplied a significant fraction of the carbon within the fatty acid and phospholipid pools. ACSS2 expression is upregulated under metabolically stressed conditions and ACSS2 silencing reduced the growth of tumor xenografts. ACSS2 exhibits copy-number gain in human breast tumors, and ACSS2 expression correlates with disease progression. These results signify a critical role for acetate consumption in the production of lipid biomass within the harsh tumor microenvironment., Graphical Abstract, Highlights • ACSS2 expression positively correlates with tumor stage and patient survival • Hypoxia and low lipid availability synergistically stimulate ACSS2 expression • Acetate is a major source of carbon for lipid synthesis during metabolic stress • ACSS2 is required for growth of tumor xenografts harboring ACSS2 copy-number gains, Schug et al. show that ACSS2 expression is increased in cancer cells under metabolic stress, and it is critical for cancer cells to use acetate as a nutritional source for lipid biomass production under this condition. Importantly, the ACSS2 expression level correlates with breast cancer progression.
- Published
- 2015
24. The Dynamics of TGF-β Signaling Are Dictated by Receptor Trafficking via the ESCRT Machinery
- Author
-
Probir Chakravarty, Rebecca E. Saunders, Ilaria Gori, Debipriya Das, Caroline S. Hill, Ming Jiang, Michael Howell, Daniel S. J. Miller, and Robert D. Bloxham
- Subjects
0301 basic medicine ,Small interfering RNA ,Epithelial-Mesenchymal Transition ,Endosome ,Ubiquitin-Protein Ligases ,Down-Regulation ,macromolecular substances ,Smad2 Protein ,General Biochemistry, Genetics and Molecular Biology ,ESCRT ,Cell Line ,03 medical and health sciences ,Mice ,Downregulation and upregulation ,Cell surface receptor ,Transforming Growth Factor beta ,Neoplasms ,Animals ,Humans ,Phosphorylation ,Receptor ,lcsh:QH301-705.5 ,Endosomal Sorting Complexes Required for Transport ,Chemistry ,Genome, Human ,Multivesicular Bodies ,Prognosis ,Survival Analysis ,Cell biology ,Activins ,Up-Regulation ,Protein Transport ,030104 developmental biology ,lcsh:Biology (General) ,Bone Morphogenetic Proteins ,Proteolysis ,Signal transduction ,Lysosomes ,Receptors, Transforming Growth Factor beta ,Transforming growth factor ,Signal Transduction - Abstract
Summary: Signal transduction pathways stimulated by secreted growth factors are tightly regulated at multiple levels between the cell surface and the nucleus. The trafficking of cell surface receptors is emerging as a key step for regulating appropriate cellular responses, with perturbations in this process contributing to human diseases, including cancer. For receptors recognizing ligands of the transforming growth factor β (TGF-β) family, little is known about how trafficking is regulated or how this shapes signaling dynamics. Here, using whole genome small interfering RNA (siRNA) screens, we have identified the ESCRT (endosomal sorting complex required for transport) machinery as a crucial determinant of signal duration. Downregulation of ESCRT components increases the outputs of TGF-β signaling and sensitizes cells to low doses of ligand in their microenvironment. This sensitization drives an epithelial-to-mesenchymal transition (EMT) in response to low doses of ligand, and we demonstrate a link between downregulation of the ESCRT machinery and cancer survival. : Miller et al. demonstrate, using whole genome siRNA screening, that TGF-β receptors are targeted for degradation by the ESCRT machinery. Inhibiting ESCRT components upregulates long-term TGF-β signaling and enhances functional outputs of the pathway to sensitize cells to low levels of ligand in the micro-environment. Keywords: epithelial-to-mesenchymal transition, ESCRT machinery, receptor trafficking, signaling dynamics, SMAD2, TGF-β
- Published
- 2017
25. Actin and Src-family kinases regulate nuclear YAP1 and its export
- Author
-
Anna M Dowbaj, Michael Howell, Erik Sahai, Ming Jiang, Robert P. Jenkins, and Nil Ege
- Subjects
YAP1 ,XPO1 ,Chemistry ,Phosphorylation ,Nuclear transport ,Nuclear export signal ,Cytoskeleton ,Nuclear localization sequence ,Proto-oncogene tyrosine-protein kinase Src ,Cell biology - Abstract
SummaryThe transcriptional regulator YAP1 is critical for the pathological activation of fibroblasts. In normal fibroblasts YAP1 is predominantly located in the cytoplasm, while in activated cancer-associated fibroblasts it exhibits nuclear localization and promotes the expression of many genes required for pro-tumorigenic functions. Here, we investigate the dynamics of YAP1 shuttling in normal and activated fibroblasts, using EYFP-YAP1, quantitative photo-bleaching methods, and mathematical modeling. We find that both 14-3-3 and TEAD binding modulate YAP1 shuttling, but neither affects nuclear import. Instead, we find that YAP1 serine phosphorylation is required for nuclear export. Furthermore, YAP1 nuclear accumulation in activated fibroblasts results from Src and actomyosin-dependent suppression of phosphorylated YAP1 export. Finally, we show that nuclear constrained YAP1, upon XPO1 depletion, remains sensitive to blockade of actomyosin function. Together, these data place nuclear export at the center of YAP1 regulation and indicate that the cytoskeleton can regulate YAP1 within the nucleus.
- Published
- 2017
- Full Text
- View/download PDF
26. Actomyosin drives cancer cell nuclear dysmorphia and threatens genome stability
- Author
-
Marco Montagner, Karoly Szuhai, Murielle P. Serres, Matthew R. G. Russell, Mark Petronczki, Tohru Takaki, Simon J. Boulton, Michael Howell, Lucy M. Collinson, Erik Sahai, and Maël Le Berre
- Subjects
0301 basic medicine ,Genome instability ,Cell Nucleus Shape ,DNA damage ,Nuclear Envelope ,Science ,General Physics and Astronomy ,macromolecular substances ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Genomic Instability ,Article ,Contractility ,03 medical and health sciences ,Myosin-Light-Chain Phosphatase ,Neoplasms ,Protein Phosphatase 1 ,medicine ,Humans ,Actin ,Genetics ,Multidisciplinary ,General Chemistry ,Actomyosin ,Cell biology ,Cell nucleus ,030104 developmental biology ,medicine.anatomical_structure ,Cancer cell ,Myosin-light-chain phosphatase ,HeLa Cells - Abstract
Altered nuclear shape is a defining feature of cancer cells. The mechanisms underlying nuclear dysmorphia in cancer remain poorly understood. Here we identify PPP1R12A and PPP1CB, two subunits of the myosin phosphatase complex that antagonizes actomyosin contractility, as proteins safeguarding nuclear integrity. Loss of PPP1R12A or PPP1CB causes nuclear fragmentation, nuclear envelope rupture, nuclear compartment breakdown and genome instability. Pharmacological or genetic inhibition of actomyosin contractility restores nuclear architecture and genome integrity in cells lacking PPP1R12A or PPP1CB. We detect actin filaments at nuclear envelope rupture sites and define the Rho-ROCK pathway as the driver of nuclear damage. Lamin A protects nuclei from the impact of actomyosin activity. Blocking contractility increases nuclear circularity in cultured cancer cells and suppresses deformations of xenograft nuclei in vivo. We conclude that actomyosin contractility is a major determinant of nuclear shape and that unrestrained contractility causes nuclear dysmorphia, nuclear envelope rupture and genome instability., Recent findings suggest that forces acting on the cell nucleus can cause DNA damage, but the mechanisms are unclear. Here Takaki et al. report that actomyosin is a determinant of nuclear shape and that unrestrained contractility elicits nuclear envelope rupture and genome instability in cancer cells.
- Published
- 2017
27. Quantitative Analysis Reveals that Actin and Src-Family Kinases Regulate Nuclear YAP1 and Its Export
- Author
-
Erik Sahai, Charles T. Foster, Ming Jiang, Michael Howell, Robert P. Jenkins, Anna M Dowbaj, Nil Ege, and Steven Hooper
- Subjects
0301 basic medicine ,Cytoplasm ,Histology ,Active Transport, Cell Nucleus ,Cell Cycle Proteins ,YAP1 ,photobleaching ,Article ,fibroblast ,Pathology and Forensic Medicine ,03 medical and health sciences ,XPO1 ,Mice ,Cell Movement ,Cell Line, Tumor ,Animals ,Humans ,Phosphorylation ,Cytoskeleton ,Nuclear export signal ,Actin ,Src-family kinases ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Cell Nucleus ,Chemistry ,mathematical modeling ,YAP-Signaling Proteins ,Cell Biology ,Fibroblasts ,Models, Theoretical ,Phosphoproteins ,Actins ,Cell biology ,DNA-Binding Proteins ,030104 developmental biology ,Gene Expression Regulation ,actomyosin cytoskeleton ,nuclear export ,Nuclear transport ,Proto-oncogene tyrosine-protein kinase Src ,Signal Transduction - Abstract
Summary The transcriptional regulator YAP1 is critical for the pathological activation of fibroblasts. In normal fibroblasts, YAP1 is located in the cytoplasm, while in activated cancer-associated fibroblasts, it is nuclear and promotes the expression of genes required for pro-tumorigenic functions. Here, we investigate the dynamics of YAP1 shuttling in normal and activated fibroblasts, using EYFP-YAP1, quantitative photobleaching methods, and mathematical modeling. Imaging of migrating fibroblasts reveals the tight temporal coupling of cell shape change and altered YAP1 localization. Both 14-3-3 and TEAD binding modulate YAP1 shuttling, but neither affects nuclear import. Instead, we find that YAP1 nuclear accumulation in activated fibroblasts results from Src and actomyosin-dependent suppression of phosphorylated YAP1 export. Finally, we show that nuclear-constrained YAP1, upon XPO1 depletion, remains sensitive to blockade of actomyosin function. Together, these data place nuclear export at the center of YAP1 regulation and indicate that the cytoskeleton can regulate YAP1 within the nucleus., Graphical Abstract, Highlights • Photobleaching coupled with mathematical modeling identifies YAP1 dynamics • Regulation of nuclear export is key determinant of YAP1 localization • Serine phosphorylation is required for YAP1 nuclear export through XPO1 • Nuclear YAP1 remains sensitive to actin and Src-family kinase regulation, Ege et al. use quantitative analysis of photobleaching experiments to reveal that nuclear export is a convergent point of YAP1 regulation by actomyosin, Src-family kinases, and LATS1/2. Further, actomyosin and Src-family kinases additionally regulate YAP1 transcription competence via tyrosine phosphorylation.
- Published
- 2017
28. Salt-inducible kinases regulate growth through the Hippo signalling pathway in Drosophila
- Author
-
Maxine V. Holder, Moritz J. Rossner, Ming Jiang, Ieva Gailite, Tobias M. Maile, Elena Ciirdaeva, Michael C. Wehr, Rachael Instrell, Michael Howell, Rebecca E. Saunders, and Nicolas Tapon
- Subjects
endocrine system ,animal structures ,Hpo signalling ,proliferation ,Cell Cycle Proteins ,Protein Serine-Threonine Kinases ,Biology ,Article ,Cell Line ,Protein Interaction Mapping ,Animals ,Drosophila Proteins ,Wings, Animal ,Phosphorylation ,Sik3 ,Sik2 ,Hippo signaling pathway ,Protein-Serine-Threonine Kinases ,split TEV ,Kinase ,fungi ,Intracellular Signaling Peptides and Proteins ,Nuclear Proteins ,AMPK ,YAP-Signaling Proteins ,Organ Size ,Cell Biology ,Hedgehog signaling pathway ,Cell biology ,body regions ,Drosophila melanogaster ,14-3-3 Proteins ,Gene Expression Regulation ,Gene Knockdown Techniques ,Trans-Activators ,RNAi screening ,RNA Interference ,sense organs ,Signal transduction ,Protein Processing, Post-Translational ,Drosophila Protein ,Protein Binding ,Signal Transduction - Abstract
The specification of tissue size during development involves the coordinated action of many signalling pathways responding to organ-intrinsic signals, such as morphogen gradients, and systemic cues, such as nutrient status. The conserved Hippo (Hpo) pathway, which promotes both cell cycle exit and apoptosis, is a major determinant of size control.The pathway core is a kinase cassette, comprising the kinases Hpo and Warts (Wts) and the scaffold proteins Salvador (Sav) and Mats, which inactivates the pro-growth transcriptional co-activator Yorkie (Yki). We performed a split TEV-based genome-wide RNAi screen for modulators of Hpo signalling. We characterise the Drosophila salt-inducible kinases (Sik2 and Sik3) as negative regulators of Hpo signalling. Activated Sik kinases increase Yki target expression and promote tissue overgrowth through phosphorylation of Sav at Ser413. Since Sik kinases have been implicated in nutrient sensing, this suggests a link between the Hippo pathway and systemic growth control.
- Published
- 2012
29. Mice develop normally in the absence of Smad4 nucleocytoplasmic shuttling
- Author
-
Caroline S. Hill, Michael Howell, Christine A. Biondi, Elizabeth K. Bikoff, Ayesha Islam, Elizabeth J. Robertson, and Debipriya Das
- Subjects
Transcriptional Activation ,Cytoplasm ,animal structures ,RNA Splicing ,SMAD ,Biology ,Bone morphogenetic protein ,Biochemistry ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Transforming Growth Factor beta ,Animals ,Nuclear export signal ,Molecular Biology ,Alleles ,Cells, Cultured ,DNA Primers ,Smad4 Protein ,030304 developmental biology ,Cell Nucleus ,Mice, Knockout ,0303 health sciences ,Base Sequence ,integumentary system ,Gene targeting ,Cell Biology ,Embryonic stem cell ,Molecular biology ,Mice, Mutant Strains ,digestive system diseases ,Gene Targeting ,embryonic structures ,Chromosomal region ,biological phenomena, cell phenomena, and immunity ,030217 neurology & neurosurgery ,Nuclear localization sequence ,Signal Transduction ,Research Article - Abstract
Smad4 in partnership with R-Smads (receptor-regulated Smads) activates TGF-β (transforming growth factor-β)-dependent signalling pathways essential for early mouse development. Smad4 null embryos die shortly after implantation due to severe defects in cell proliferation and visceral endoderm differentiation. In the basal state, Smad4 undergoes continuous shuttling between the cytoplasm and the nucleus due to the combined activities of an N-terminal NLS (nuclear localization signal) and an NES (nuclear export signal) located in its linker region. Cell culture experiments suggest that Smad4 nucleocytoplasmic shuttling plays an important role in TGF-β signalling. In the present study we have investigated the role of Smad4 shuttling in vivo using gene targeting to engineer two independent mutations designed to eliminate Smad4 nuclear export. As predicted this results in increased levels of Smad4 in the nucleus of homozygous ES cells (embryonic stem cells) and primary keratinocytes, in the presence or absence of ligand. Neither mutation affects Smad4 expression levels nor its ability to mediate transcriptional activation in homozygous cell lines. Remarkably mouse mutants lacking the Smad4 NES develop normally. Smad4 NES mutants carrying one copy of a Smad4 null allele also fail to display developmental defects. The present study clearly demonstrates that Smad4 nucleocytoplasmic shuttling is not required for embryonic development or tissue homoeostasis in normal, healthy adult mice. Abbreviations: BMP, bone morphogenetic proteins; CRM1, chromosomal region maintenance 1; d.p.c., days post-coitum; ES cell, embryonic stem cell; FBS, foetal bovine serum; GDF, growth and differentiation factor; LMB, leptomycin B; MEF, murine embryonic fibroblast; MH domain, Mad homology domain; NES, nuclear export signal; NLS, nuclear localization signal; RPA, ribonuclease protection assay; R-Smad, receptor-regulated Smad; SnoN, Ski-related novel protein N; TGF-β, transforming growth factor-β
- Published
- 2016
30. The Microtubule-Associated Rho Activating Factor GEF-H1 Interacts with Exocyst Complex to Regulate Vesicle Traffic
- Author
-
Michael A. White, Ritu Pathak, Michael Howell, Céline DerMardirossian, Violaine D. Delorme-Walker, Gary M. Bokoch, and Anthony Anselmo
- Subjects
RHOA ,Exocyst ,Biology ,Microtubules ,Article ,Exocytosis ,General Biochemistry, Genetics and Molecular Biology ,Microscopy, Electron, Transmission ,Microtubule ,Guanine Nucleotide Exchange Factors ,Humans ,Molecular Biology ,Vesicle ,Biological Transport ,Cell Biology ,Exocyst assembly ,Cell biology ,Enzyme Activation ,biology.protein ,Guanine nucleotide exchange factor ,Signal transduction ,rhoA GTP-Binding Protein ,Rho Guanine Nucleotide Exchange Factors ,HeLa Cells ,Protein Binding ,Signal Transduction ,Developmental Biology - Abstract
SummaryThe exocyst complex plays a critical role in targeting and tethering vesicles to specific sites of the plasma membrane. These events are crucial for polarized delivery of membrane components to the cell surface, which is critical for cell motility and division. Though Rho GTPases are involved in regulating actin dynamics and membrane trafficking, their role in exocyst-mediated vesicle targeting is not very clear. Herein, we present evidence that depletion of GEF-H1, a guanine nucleotide exchange factor for Rho proteins, affects vesicle trafficking. Interestingly, we found that GEF-H1 directly binds to exocyst component Sec5 in a Ral GTPase-dependent manner. This interaction promotes RhoA activation, which then regulates exocyst assembly/localization and exocytosis. Taken together, our work defines a mechanism for RhoA activation in response to RalA-Sec5 signaling and involvement of GEF-H1/RhoA pathway in the regulation of vesicle trafficking.
- Published
- 2012
31. Determination of synthetic lethal interactions in KRAS oncogene-dependent cancer cells reveals novel therapeutic targeting strategies
- Author
-
Britta Weigelt, Julian Downward, Michael Howell, Patricia H. Warne, Michael Steckel, Becky Saunders, David C. Hancock, Miriam Molina-Arcas, Michaela Marani, Hanna Kuznetsov, and Gavin Kelly
- Subjects
Mutant ,Apoptosis ,Cell Cycle Proteins ,Synthetic lethality ,medicine.disease_cause ,Deoxycytidine ,Bortezomib ,RNA, Small Interfering ,topoisomerase ,Mutation ,GATA2 ,Nuclear Proteins ,Boronic Acids ,GATA2 Transcription Factor ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,DNA Topoisomerases, Type I ,Gene Knockdown Techniques ,Pyrazines ,Colonic Neoplasms ,Original Article ,RNA Interference ,KRAS ,Proteasome Inhibitors ,Transcriptional Activation ,oncogene addiction ,Proteasome Endopeptidase Complex ,Cell Survival ,Antineoplastic Agents ,Biology ,Proto-Oncogene Proteins p21(ras) ,Cell Line, Tumor ,Proto-Oncogene Proteins ,medicine ,Humans ,Molecular Biology ,Alleles ,Oncogene ,Cancer ,Cell Biology ,medicine.disease ,Gemcitabine ,proteasome ,synthetic lethal ,Cancer cell ,Cancer research ,ras Proteins ,Topoisomerase I Inhibitors ,Topotecan - Abstract
Oncogenic mutations in RAS genes are very common in human cancer, resulting in cells with well-characterized selective advantages, but also less well-understood vulnerabilities. We have carried out a large-scale loss-of-function screen to identify genes that are required by KRAS-transformed colon cancer cells, but not by derivatives lacking this oncogene. Top-scoring genes were then tested in a larger panel of KRAS mutant and wild-type cancer cells. Cancer cells expressing oncogenic KRAS were found to be highly dependent on the transcription factor GATA2 and the DNA replication initiation regulator CDC6. Extending this analysis using a collection of drugs with known targets, we found that cancer cells with mutant KRAS showed selective addiction to proteasome function, as well as synthetic lethality with topoisomerase inhibition. Combination targeting of these functions caused improved killing of KRAS mutant cells relative to wild-type cells. These observations suggest novel targets and new ways of combining existing therapies for optimal effect in RAS mutant cancers, which are traditionally seen as being highly refractory to therapy.
- Published
- 2012
32. Genome-wide siRNA screen reveals amino acid starvation-induced autophagy requires SCOC and WAC
- Author
-
Nicole C. McKnight, Rebecca E. Saunders, Sharon A. Tooze, Terje Johansen, Endalkachew Ashenafi Alemu, Michael Howell, and Harold B. J. Jefferies
- Subjects
chemistry.chemical_classification ,General Immunology and Microbiology ,General Neuroscience ,Autophagy ,UVRAG ,ULK1 ,Biology ,BAG3 ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,Amino acid ,chemistry ,Biochemistry ,Cytoplasm ,Molecular Biology ,Gene ,FEZ1 - Abstract
Autophagy is a catabolic process by which cytoplasmic components are sequestered and transported by autophagosomes to lysosomes for degradation, enabling recycling of these components and providing cells with amino acids during starvation. It is a highly regulated process and its deregulation contributes to multiple diseases. Despite its importance in cell homeostasis, autophagy is not fully understood. To find new proteins that modulate starvation-induced autophagy, we performed a genome-wide siRNA screen in a stable human cell line expressing GFP–LC3, the marker-protein for autophagosomes. Using stringent validation criteria, our screen identified nine novel autophagy regulators. Among the hits required for autophagosome formation are SCOC (short coiled-coil protein), a Golgi protein, which interacts with fasciculation and elongation protein zeta 1 (FEZ1), an ULK1-binding protein. SCOC forms a starvation-sensitive trimeric complex with UVRAG (UV radiation resistance associated gene) and FEZ1 and may regulate ULK1 and Beclin 1 complex activities. A second candidate WAC is required for starvation-induced autophagy but also acts as a potential negative regulator of the ubiquitin-proteasome system. The identification of these novel regulatory proteins with diverse functions in autophagy contributes towards a fuller understanding of autophagosome formation.
- Published
- 2012
33. Nuclear import impairment causes cytoplasmic trans-activation response DNA-binding protein accumulation and is associated with frontotemporal lobar degeneration
- Author
-
Pietro Fratta, Jean-Marc Gallo, Boris Rogelj, Claire Troakes, Zupunski, Agnes L. Nishimura, Tibor Hortobágyi, Christopher Shaw, Michael Howell, and Claudia Kathe
- Subjects
Male ,alpha Karyopherins ,Cytoplasm ,Pathology ,medicine.medical_specialty ,Active Transport, Cell Nucleus ,Cell Line ,Mice ,Ubiquitin ,Cellular Apoptosis Susceptibility Protein ,Cell Line, Tumor ,mental disorders ,medicine ,Animals ,Humans ,Elméleti orvostudományok ,Amyotrophic lateral sclerosis ,Aged ,Glutathione Transferase ,biology ,Amyotrophic Lateral Sclerosis ,Brain ,nutritional and metabolic diseases ,Orvostudományok ,Frontotemporal lobar degeneration ,Middle Aged ,beta Karyopherins ,medicine.disease ,nervous system diseases ,Cell biology ,DNA-Binding Proteins ,Cell nucleus ,medicine.anatomical_structure ,Spinal Cord ,biology.protein ,Female ,Neurology (clinical) ,Frontotemporal Lobar Degeneration ,Nuclear transport ,Nuclear localization sequence ,Cellular apoptosis susceptibility protein ,Signal Transduction - Abstract
Trans-activation response DNA-binding protein (TDP-43) accumulation is the major component of ubiquitinated protein inclusions found in patients with amyotrophic lateral sclerosis, and frontotemporal lobar degeneration with TDP-43 positive ubiquitinated inclusions, recently relabelled the 'TDP-43 proteinopathies'. TDP-43 is predominantly located in the nucleus, however, in disease it mislocalizes to the cytoplasm where it aggregates to form hallmark pathological inclusions. The identification of TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis cases confirms its pathogenic role; but it is wild-type TDP-43 that is deposited in the vast majority of TDP-43 proteinopathies, implicating other unknown factors for its mislocalization and aggregation. One such mechanism may be defective nuclear import of TDP-43 protein, as a disruption of its nuclear localization signal leads to mislocalization and aggregation of TDP-43 in the cytoplasm. In order to explore the factors that regulate the nuclear import of TDP-43, we used a small interfering RNA library to silence 82 proteins involved in nuclear transport and found that knockdowns of karyopherin-beta1 and cellular apoptosis susceptibility protein resulted in marked cytoplasmic accumulation of TDP-43. In glutathione S-transferase pull-down assays, TDP-43 bound to karyopherin-alphas, thereby confirming the classical nuclear import pathway for the import of TDP-43. Analysis of the expression of chosen nuclear import factors in post-mortem brain samples from patients with TDP-43 positive frontotemporal lobar degeneration, and spinal cord samples from patients with amyotrophic lateral sclerosis, revealed a considerable reduction in expression of cellular apoptosis susceptibility protein in frontotemporal lobar degeneration. We propose that cellular apoptosis susceptibility protein associated defective nuclear transport may play a mechanistic role in the pathogenesis of the TDP-43 positive frontotemporal lobar degeneration.
- Published
- 2010
34. Two highly related regulatory subunits of PP2A exert opposite effects on TGF-β/Activin/Nodal signalling
- Author
-
Laurel A. Raftery, Jing Cao, Michael Howell, Bernhard Schmierer, Julie Batut, and Caroline S. Hill
- Subjects
medicine.medical_specialty ,Embryo, Nonmammalian ,animal structures ,Nodal Protein ,Activin Receptors ,Xenopus ,Protein subunit ,Receptor, Transforming Growth Factor-beta Type I ,Smad2 Protein ,Protein Serine-Threonine Kinases ,Xenopus Proteins ,Article ,Cell Line ,Mice ,Transforming Growth Factor beta ,Internal medicine ,Phosphoprotein Phosphatases ,medicine ,Animals ,Drosophila Proteins ,Humans ,Wings, Animal ,Protein Phosphatase 2 ,Phosphorylation ,Molecular Biology ,Activin type 2 receptors ,biology ,Gene Expression Regulation, Developmental ,Transforming growth factor beta ,Activin receptor ,Protein phosphatase 2 ,Activins ,Cell biology ,Protein Subunits ,Phenotype ,Endocrinology ,embryonic structures ,biology.protein ,Drosophila ,NODAL ,Activin Receptors, Type I ,Receptors, Transforming Growth Factor beta ,ACVR2B ,Signal Transduction ,Developmental Biology - Abstract
We identify Balpha (PPP2R2A) and Bdelta (PPP2R2D), two highly related members of the B family of regulatory subunits of the protein phosphatase PP2A, as important modulators of TGF-beta/Activin/Nodal signalling that affect the pathway in opposite ways. Knockdown of Balpha in Xenopus embryos or mammalian tissue culture cells suppresses TGF-beta/Activin/Nodal-dependent responses, whereas knockdown of Bdelta enhances these responses. Moreover, in Drosophila, overexpression of Smad2 rescues a severe wing phenotype caused by overexpression of the single Drosophila PP2A B subunit Twins. We show that, in vertebrates, Balpha enhances TGF-beta/Activin/Nodal signalling by stabilising the basal levels of type I receptor, whereas Bdelta negatively modulates these pathways by restricting receptor activity. Thus, these highly related members of the same subfamily of PP2A regulatory subunits differentially regulate TGF-beta/Activin/Nodal signalling to elicit opposing biological outcomes.
- Published
- 2008
35. Kinesin-Mediated Transport of Smad2 Is Required for Signaling in Response to TGF-β Ligands
- Author
-
Michael Howell, Caroline S. Hill, and Julie Batut
- Subjects
Embryo, Nonmammalian ,animal structures ,Nodal Protein ,Recombinant Fusion Proteins ,Xenopus ,Protein subunit ,Green Fluorescent Proteins ,Kinesins ,Nodal signaling ,Smad2 Protein ,macromolecular substances ,Biology ,Ligands ,Microtubules ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Transforming Growth Factor beta ,Microtubule ,medicine ,Animals ,Humans ,Phosphorylation ,Molecular Biology ,Zebrafish ,Cell Nucleus ,Cell Biology ,biology.organism_classification ,Activins ,Cell biology ,Protein Transport ,medicine.anatomical_structure ,Biochemistry ,Cytoplasm ,embryonic structures ,NIH 3T3 Cells ,Kinesin ,NODAL ,Microtubule-Associated Proteins ,Nucleus ,Protein Binding ,Signal Transduction ,Developmental Biology - Abstract
Summary During vertebrate development, Activin/Nodal-related ligands signal through Smad2, leading to its activation and accumulation in the nucleus. Here, we demonstrate that Smad2 constantly shuttles between the cytoplasm and nucleus both in early Xenopus embryo explants and in living zebrafish embryos, providing a mechanism whereby the intracellular components of the pathway constantly monitor receptor activity. We have gone on to demonstrate that an intact microtubule network and kinesin ATPase activity are required for Smad2 phosphorylation and nuclear accumulation in response to Activin/Nodal in early vertebrate embryos and TGF-β in mammalian cells. The kinesin involved is kinesin-1, and Smad2 interacts with the kinesin-1 light chain subunit. Interfering with kinesin activity in Xenopus and zebrafish embryos phenocopies loss of Nodal signaling. Our results reveal that kinesin-mediated transport of Smad2 along microtubules to the receptors is an essential step in ligand-induced Smad2 activation.
- Published
- 2007
- Full Text
- View/download PDF
36. The sorting protein PACS-2 promotes ErbB signalling by regulating recycling of the metalloproteinase ADAM17
- Author
-
Rachael Instrell, Anne-Mette Bornhardt Hedegaard, Camilla Fröhlich, Camilla Hansson Petersen, Peter J. Parker, Gary Thomas, Marie Kveiborg, Sylvain Auclair, Katelyn M. Atkins, Sarah Louise Dombernowsky, Reidar Albrechtsen, Kasper Johansen Mygind, Laurel Thomas, Michael Howell, and Jacob Samsøe-Petersen
- Subjects
Endosome ,Vesicular Transport Proteins ,Regulator ,General Physics and Astronomy ,ADAM17 Protein ,Biology ,Article ,Gene Expression Regulation, Enzymologic ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,ErbB ,Cell Line, Tumor ,Animals ,Humans ,030304 developmental biology ,0303 health sciences ,Metalloproteinase ,Gene knockdown ,Multidisciplinary ,Oncogene Proteins v-erbB ,General Chemistry ,Cell biology ,ADAM Proteins ,030220 oncology & carcinogenesis ,Signal transduction ,Genome-Wide Association Study ,Signal Transduction - Abstract
The metalloproteinase ADAM17 activates ErbB signalling by releasing ligands from the cell surface, a key step underlying epithelial development, growth, and tumour progression. However, mechanisms acutely controlling ADAM17 cell-surface availability to modulate the extent of ErbB ligand release are poorly understood. Here, through a functional genome-wide siRNA screen, we identify the sorting protein PACS-2 as a regulator of ADAM17 trafficking and ErbB signalling. PACS-2 loss reduces ADAM17 cell-surface levels and ADAM17-dependent ErbB ligand shedding, without apparent effects on related proteases. PACS-2 co-localizes with ADAM17 on early endosomes and PACS-2 knockdown decreases the recycling and stability of internalized ADAM17. Hence, PACS-2 sustains ADAM17 cell-surface activity by diverting ADAM17 away from degradative pathways. Interestingly, Pacs2-deficient mice display significantly reduced levels of phosphorylated EGFR and intestinal proliferation. We suggest that this mechanism controlling ADAM17 cell-surface availability and EGFR signalling may play a role in intestinal homeostasis, with potential implications for cancer biology.
- Published
- 2015
37. Ccdc13 is a novel human centriolar satellite protein required for ciliogenesis and genome stability
- Author
-
Sarah L. Maslen, Alvin J.X. Lee, Christopher J. Staples, Ryan D. Beveridge, Spencer J. Collis, Katie N. Myers, Abhijit A. Patil, Anna E. Howard, Simon J. Boulton, Giancarlo Barone, J. Mark Skehel, Charles Swanton, and Michael Howell
- Subjects
Genome instability ,Centriole ,Cell Cycle Proteins ,Cell Biology ,Biology ,medicine.disease ,HCT116 Cells ,Transfection ,Genomic Instability ,Cell biology ,Ciliopathy ,PCM1 ,HEK293 Cells ,Microtubule ,Ciliogenesis ,medicine ,Basal body ,Humans ,Centriolar satellite ,Cilia ,Centrioles ,HeLa Cells - Abstract
Here, we identify coiled-coil domain-containing protein 13 (Ccdc13) in a genome-wide RNA interference screen for regulators of genome stability. We establish that Ccdc13 is a newly identified centriolar satellite protein that interacts with PCM1, Cep290 and pericentrin and prevents the accumulation of DNA damage during mitotic transit. Depletion of Ccdc13 results in the loss of microtubule organisation in a manner similar to PCM1 and Cep290 depletion, although Ccdc13 is not required for satellite integrity. We show that microtubule regrowth is enhanced in Ccdc13-depleted cells, but slowed in cells that overexpress Ccdc13. Furthermore, in serum-starved cells, Ccdc13 localises to the basal body, is required for primary cilia formation and promotes the localisation of the ciliopathy protein BBS4 to both centriolar satellites and cilia. These data highlight the emerging link between DNA damage response factors, centriolar and peri-centriolar satellites and cilia-associated proteins and implicate Ccdc13 as a centriolar satellite protein that functions to promote both genome stability and cilia formation.
- Published
- 2014
38. Loss of Smad4 Function in Pancreatic Tumors
- Author
-
Robb E. Wilentz, Michael Howell, Michael John Owen, Caroline S. Hill, Diane Maurice, and Christophe E. Pierreux
- Subjects
Genetics ,Nonsense mutation ,Mutant ,Wild type ,Cell Biology ,Biology ,medicine.disease_cause ,Biochemistry ,Cell biology ,Mutant protein ,medicine ,Homomeric ,Smad2 Protein ,Carcinogenesis ,Molecular Biology ,Transcription factor - Abstract
At early stages of tumorigenesis, the transforming growth factor-beta (TGF-beta) signaling pathway is thought to have tumor suppressor activity as a result of its ability to arrest the growth of epithelial cells. Smad4 plays a pivotal role in the TGF-beta signaling pathway and has been identified as a tumor suppressor, being mutated or deleted in approximately 50% of pancreatic carcinomas and 15% of colorectal cancers. A nonsense mutation generating a C-terminal truncation of 38 amino acids in the Smad4 protein has been identified in a pancreatic adenocarcinoma (Hahn, S. A., Schutte, M., Hoque, A. T., Moskaluk, C. A., da Costa, L. T., Rozenblum, E., Weinstein, C. L., Fischer, A., Yeo, C. J., Hruban, R. H., and Kern, S. E. (1996) Science 271, 350-353), and here we investigate the functional consequences of this mutation. We demonstrate that the C-terminal truncation prevents Smad4 homomeric complex formation and heteromeric complex formation with activated Smad2. Furthermore, the mutant protein is unable to be recruited to DNA by transcription factors and hence cannot form transcriptionally active DNA-binding complexes. These observations are supported by molecular modeling, which indicates that the truncation removes residues critical for homomeric and heteromeric Smad complex formation. We go on to show that the mutant Smad4 is highly unstable compared with wild type Smad4 and is rapidly degraded through the ubiquitin-proteasome pathway. Consistent with this, we demonstrate that the pancreatic adenocarcinoma harboring this mutated allele, in conjunction with loss of the other allele, expresses no Smad4 protein. Thus we conclude that these tumors completely lack Smad4 activity.
- Published
- 2001
39. New B-type cyclin synthesis is required between meiosis I and II duringXenopusoocyte maturation
- Author
-
Muhammad Sohail, Tod Duncan, Andrea Klotzbücher, Kate Fletcher, Jane Kirk, Michael Howell, Tim Hunt, Katherine le Guellec, and Helfrid Hochegger
- Subjects
Embryo, Nonmammalian ,Maturation-Promoting Factor ,Molecular Sequence Data ,Maturation promoting factor ,Xenopus ,Cyclin B ,Xenopus Proteins ,Cycloheximide ,Xenopus laevis ,chemistry.chemical_compound ,Meiosis ,Cyclins ,medicine ,Animals ,Amino Acid Sequence ,Molecular Biology ,Cyclin ,biology ,urogenital system ,Meiosis II ,Gene Expression Regulation, Developmental ,Oligonucleotides, Antisense ,Oocyte ,biology.organism_classification ,Cell biology ,medicine.anatomical_structure ,chemistry ,Oocytes ,biology.protein ,Female ,Cyclin A2 ,Developmental Biology - Abstract
Progression through meiosis requires two waves of maturation promoting factor (MPF) activity corresponding to meiosis I and meiosis II. Frog oocytes contain a pool of inactive ‘pre-MPF’ consisting of cyclin-dependent kinase 1 bound to B-type cyclins, of which we now find three previously unsuspected members, cyclins B3, B4 and B5. Protein synthesis is required to activate pre-MPF, and we show here that this does not require new B-type cyclin synthesis, probably because of a large maternal stockpile of cyclins B2 and B5. This stockpile is degraded after meiosis I and consequently, the activation of MPF for meiosis II requires new cyclin synthesis, principally of cyclins B1 and B4, whose translation is strongly activated after meiosis I. If this wave of new cyclin synthesis is ablated by antisense oligonucleotides, the oocytes degenerate and fail to form a second meiotic spindle. The effects on meiotic progression are even more severe when all new protein synthesis is blocked by cycloheximide added after meiosis I, but can be rescued by injection of indestructible B-type cyclins. B-type cyclins and MPF activity are required to maintain c-mos and MAP kinase activity during meiosis II, and to establish the metaphase arrest at the end of meiotic maturation. We discuss the interdependence of c-mos and MPF, and reveal an important role for translational control of cyclin synthesis between the two meiotic divisions.
- Published
- 2001
40. Interaction between the Origin Recognition Complex and the Replication Licensing Systemin Xenopus
- Author
-
Michael Howell, Alison Rowles, Gerard I. Evan, James P. J. Chong, J. Julian Blow, and Lamorna Brown
- Subjects
DNA Replication ,Male ,Molecular Sequence Data ,Origin Recognition Complex ,Eukaryotic DNA replication ,Xenopus Proteins ,Pre-replication complex ,General Biochemistry, Genetics and Molecular Biology ,DNA replication factor CDT1 ,Xenopus laevis ,ORC6 ,Control of chromosome duplication ,Minichromosome maintenance ,Animals ,Humans ,Amino Acid Sequence ,Cloning, Molecular ,Base Sequence ,Cell-Free System ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,Cell Cycle ,Spermatozoa ,Molecular biology ,Chromatin ,Cell biology ,DNA-Binding Proteins ,Licensing factor ,Immunologic Techniques ,Oocytes ,biology.protein ,Origin recognition complex ,Sequence Alignment - Abstract
The origin recognition complex (ORC) binds to origins of replication in budding yeast. We have cloned a Xenopus homolog of the largest ORC polypeptide (XORC1). Immunodepletion of XOrc1 from Xenopus egg extracts blocks the initiation of DNA replication. We have purified Xenopus ORC, consisting of a protein complex similar to yeast ORC. In Xenopus egg extracts, ORC associates with chromatin throughout G1 and S phases. RLF-M, a component of the replication licensing system, also associates with chromatin early in the cell cycle but dissociates during S phase. We show that the assembly of RLF-M onto chromatin is dependent on the presence of chromatin-bound ORC, leading to sequential assembly of initiation proteins onto replication origins during the cell cycle.
- Published
- 1996
41. Binding of dynein intermediate chain 2 to paxillin controls focal adhesion dynamics and migration
- Author
-
David Frith, Peter J. Parker, Mark Linch, Michael Howell, Katrina Boeckeler, Majid Hafezparast, Carine Rossé, Karin Barnouin, Ali Sayed Morsi, and Simone Radtke
- Subjects
Cytoplasmic Dyneins ,Difference gel electrophoresis ,Dynein ,Molecular Sequence Data ,macromolecular substances ,Kidney ,environment and public health ,Serine ,Focal adhesion ,Cell Movement ,Cell Adhesion ,Animals ,Amino Acid Sequence ,Phosphorylation ,Paxillin ,Cells, Cultured ,biology ,Dyneins ,Cell migration ,Cell Biology ,Cell biology ,Rats ,enzymes and coenzymes (carbohydrates) ,biology.protein ,Dynactin - Abstract
In migrating NRK cells, aPKCs control the dynamics of turnover of paxillin-containing focal adhesions (FA) determining migration rate. Using a proteomic approach (two-dimensional fluorescence difference gel electrophoresis), dynein intermediate chain 2 (dynein IC2) was identified as a protein that is phosphorylated inducibly during cell migration in a PKC-regulated manner. By gene silencing and co-immunoprecipitation studies, we show that dynein IC2 regulates the speed of cell migration through its interaction with paxillin. This interaction is controlled by serine 84 phosphorylation, which lies on the aPKC pathway. The evidence presented thus links aPKC control of migration to the dynein control of FA turnover through paxillin.
- Published
- 2012
42. Genome-wide RNA interference analysis of renal carcinoma survival regulators identifies MCT4 as a Warburg effect metabolic target
- Author
-
Charles Swanton, Pierre Martinez, Karl Dykema, Gavin Kelly, Ming Jiang, Jean Jacques Patard, Gordon Stamp, Rebecca E. Saunders, Marcus Vetter, James Larkin, Marco Gerlinger, David Endesfelder, Nathalie Rioux-Leclercq, Michael Howell, Bradley Spencer-Dene, Rebecca A. Burrell, Claudio R. Santos, De Villemeur, Hervé, Barts Cancer Institute, Queen Mary University of London (QMUL), Cancer Research UK London Research Institute, Department of Medical Oncology, Royal Marsden Hospital, Service d'anatomie et cytologie pathologiques [Rennes] = Anatomy and Cytopathology [Rennes], CHU Pontchaillou [Rennes], Service d'urologie, Université Paris-Sud - Paris 11 (UP11)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Bicêtre, Cancer Institute, University College of London [London] (UCL), and Hôpital Bicêtre-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université Paris-Sud - Paris 11 (UP11)
- Subjects
Lactate transport ,renal cell carcinoma ,cancer metabolism ,MCT4 ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biology ,lactate transport ,[SDV.MHEP.UN]Life Sciences [q-bio]/Human health and pathology/Urology and Nephrology ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,RNA interference ,Gene expression ,medicine ,Gene silencing ,030304 developmental biology ,0303 health sciences ,Cell growth ,Microarray analysis techniques ,survival regulator ,therapeutic target ,medicine.disease ,Original Papers ,Warburg effect ,[SDV.MHEP.UN] Life Sciences [q-bio]/Human health and pathology/Urology and Nephrology ,3. Good health ,Cell biology ,Clear cell renal cell carcinoma ,030220 oncology & carcinogenesis ,SLC16A3 ,Cancer research - Abstract
International audience; Clear cell renal cell carcinoma (ccRCC) is the most common pathological subtype of kidney cancer. Here, we integrated an unbiased genome-wide RNA interference screen for ccRCC survival regulators with an analysis of recurrently over-expressed genes in ccRCC to identify new therapeutic targets in this disease. One of the most potent survival regulators, the monocarboxylate transporter MCT4 (SLC16A3), impaired ccRCC viability in all 8 ccRCC lines tested and was the 7th most over-expressed gene in a meta-analysis of 5 ccRCC expression datasets. MCT4-silencing impaired secretion of lactate generated through glycolysis and induced cell cycle arrest and apoptosis. Silencing MCT4 resulted in intracellular acidosis, and reduction in intracellular ATP production together with partial reversion of the Warburg effect in ccRCC cell lines. Intratumoral heterogeneity in the intensity of MCT4 protein expression was observed in primary ccRCCs. MCT4 protein expression analysis based on the highest intensity of expression in primary ccRCCs was associated with poorer relapse free survival, whereas modal intensity correlated with Fuhrman nuclear grade. Consistent with the potential selection of subclones enriched for MCT4 expression during disease progression, MCT4 expression was greater at sites of metastatic disease. These data suggest that MCT4 may serve as a novel metabolic target to reverse the Warburg effect and limit disease progression in ccRCC. Copyright © 2012 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
- Published
- 2012
43. Glioblastoma motility occurs in the absence of actin polymer
- Author
-
Andreas Panopoulos, Michael Howell, Robert L. Margolis, and Rati Fotedar
- Subjects
rac1 GTP-Binding Protein ,rho GTP-Binding Proteins ,Arp2/3 complex ,Motility ,RAC1 ,macromolecular substances ,Microtubules ,Polymerization ,03 medical and health sciences ,Actin remodeling of neurons ,0302 clinical medicine ,Microtubule ,Cell Movement ,Tumor Cells, Cultured ,Humans ,Actin-binding protein ,Pseudopodia ,Molecular Biology ,Actin ,030304 developmental biology ,0303 health sciences ,biology ,Cell Biology ,Articles ,Fibroblasts ,Actins ,Cell biology ,Cell Motility ,Mutation ,biology.protein ,Glioblastoma ,030217 neurology & neurosurgery - Abstract
Human glioblastoma cells are motile in the absence of intact actin polymers, following suppression of actin assembly by specific inhibitors. On the other hand, suppression of microtubules completely blocks motility. These results are clearly divergent from the standard model of actin-based cell motility in mammalian cells., In fibroblasts and keratocytes, motility is actin dependent, while microtubules play a secondary role, providing directional guidance. We demonstrate here that the motility of glioblastoma cells is exceptional, in that it occurs in cells depleted of assembled actin. Cells display persistent motility in the presence of actin inhibitors at concentrations sufficient to fully disassemble actin. Such actin independent motility is characterized by the extension of cell protrusions containing abundant microtubule polymers. Strikingly, glioblastoma cells exhibit no motility in the presence of microtubule inhibitors, at concentrations that disassemble labile microtubule polymers. In accord with an unconventional mode of motility, glioblastoma cells have some unusual requirements for the Rho GTPases. While Rac1 is required for lamellipodial protrusions in fibroblasts, expression of dominant negative Rac1 does not suppress glioblastoma migration. Other GTPase mutants are largely without unique effect, except dominant positive Rac1-Q61L, and rapidly cycling Rac1-F28L, which substantially suppress glioblastoma motility. We conclude that glioblastoma cells display an unprecedented mode of intrinsic motility that can occur in the absence of actin polymer, and that appears to require polymerized microtubules.
- Published
- 2011
44. SNW1 is a critical regulator of spatial BMP activity, neural plate border formation, and neural crest specification in vertebrate embryos
- Author
-
Caroline S. Hill, Mary Y. Wu, Michael Howell, and Marie-Christine Ramel
- Subjects
Embryo, Nonmammalian ,Xenopus ,Ectoderm ,Xenopus Proteins ,Bioinformatics ,Cell Biology/Cell Signaling ,Developmental Biology/Pattern Formation ,Developmental Biology/Molecular Development ,0302 clinical medicine ,Neural crest formation ,Biology (General) ,Zebrafish ,Developmental Biology/Embryology ,0303 health sciences ,Neural Plate ,Developmental Biology/Morphogenesis and Cell Biology ,General Neuroscience ,Neural crest ,Embryo ,11 Medical And Health Sciences ,Cell biology ,medicine.anatomical_structure ,Neural Crest ,Bone Morphogenetic Proteins ,embryonic structures ,Synopsis ,General Agricultural and Biological Sciences ,Neural plate ,Signal Transduction ,Research Article ,animal structures ,QH301-705.5 ,Embryonic Development ,Biology ,Bone morphogenetic protein ,General Biochemistry, Genetics and Molecular Biology ,Protein–protein interaction ,03 medical and health sciences ,Bmp signaling ,medicine ,Animals ,Cell Biology/Gene Expression ,030304 developmental biology ,Body Patterning ,Neural fold ,General Immunology and Microbiology ,Gastrulation ,Bone Morphogenetic Protein Receptors ,06 Biological Sciences ,Zebrafish Proteins ,biology.organism_classification ,Embryonic stem cell ,Molecular biology ,Neurulation ,Mesoderm formation ,Developmental Biology/Cell Differentiation ,07 Agricultural And Veterinary Sciences ,Carrier Proteins ,030217 neurology & neurosurgery ,Developmental Biology ,Transcription Factors - Abstract
In frog and fish embryos, SNW1 is a protein required for the spatio-temporal activity of BMP signaling necessary for neural plate border formation and specification of neural crest tissue., Bone morphogenetic protein (BMP) gradients provide positional information to direct cell fate specification, such as patterning of the vertebrate ectoderm into neural, neural crest, and epidermal tissues, with precise borders segregating these domains. However, little is known about how BMP activity is regulated spatially and temporally during vertebrate development to contribute to embryonic patterning, and more specifically to neural crest formation. Through a large-scale in vivo functional screen in Xenopus for neural crest fate, we identified an essential regulator of BMP activity, SNW1. SNW1 is a nuclear protein known to regulate gene expression. Using antisense morpholinos to deplete SNW1 protein in both Xenopus and zebrafish embryos, we demonstrate that dorsally expressed SNW1 is required for neural crest specification, and this is independent of mesoderm formation and gastrulation morphogenetic movements. By exploiting a combination of immunostaining for phosphorylated Smad1 in Xenopus embryos and a BMP-dependent reporter transgenic zebrafish line, we show that SNW1 regulates a specific domain of BMP activity in the dorsal ectoderm at the neural plate border at post-gastrula stages. We use double in situ hybridizations and immunofluorescence to show how this domain of BMP activity is spatially positioned relative to the neural crest domain and that of SNW1 expression. Further in vivo and in vitro assays using cell culture and tissue explants allow us to conclude that SNW1 acts upstream of the BMP receptors. Finally, we show that the requirement of SNW1 for neural crest specification is through its ability to regulate BMP activity, as we demonstrate that targeted overexpression of BMP to the neural plate border is sufficient to restore neural crest formation in Xenopus SNW1 morphants. We conclude that through its ability to regulate a specific domain of BMP activity in the vertebrate embryo, SNW1 is a critical regulator of neural plate border formation and thus neural crest specification., Author Summary A subset of cells in the ectoderm of vertebrate embryos becomes the neural crest, which contributes to the bones and cartilage of the adult face. The neural crest arises in a location between the epidermis, which becomes the future skin, and the neural plate, which becomes the future central nervous system. Through our studies in both frog and fish embryos, we have discovered that the protein SNW-domain containing protein 1 (SNW1) is absolutely essential for defining the edge of the neural plate, where neural crest forms. SNW-domain containing proteins have been implicated in a variety of nuclear activities ranging from transcriptional regulation and elongation to RNA splicing. We show that SNW1 functions upstream of bone morphogenetic protein (BMP) receptors to regulate BMP activity, and is necessary for the activity of the BMP signaling pathway at the neural plate border where the neural crest is specified. In the absence of SNW1, BMP activity is reduced in this region and neural crest cells are lost. Given that SNW1 family proteins are highly conserved from nematodes to humans, SNW1's BMP regulatory function is likely conserved in other animals.
- Published
- 2011
45. PKCα and PKCδ regulate ADAM17-mediated ectodomain shedding of heparin binding-EGF through separate pathways
- Author
-
Michael Howell, Christina Rowlands, Marie Kveiborg, Rachael Instrell, and Peter J. Parker
- Subjects
MAPK/ERK pathway ,Anatomy and Physiology ,Heparin-binding EGF-like growth factor ,lcsh:Medicine ,Signal transduction ,ERK signaling cascade ,Biochemistry ,Molecular cell biology ,Transmembrane signaling ,Basic Cancer Research ,Cluster Analysis ,ADAM17 Protein ,Membrane Receptor Signaling ,Biomacromolecule-Ligand Interactions ,Phosphorylation ,lcsh:Science ,Protein kinase signaling cascade ,Cells, Cultured ,Multidisciplinary ,Mechanisms of Signal Transduction ,Signaling cascades ,Drug Synergism ,Signaling in Selected Disciplines ,Cell biology ,ErbB Receptors ,Protein Kinase C-delta ,Oncology ,Ectodomain ,Medicine ,Intercellular Signaling Peptides and Proteins ,Tetradecanoylphorbol Acetate ,Tyrosine kinase ,Metabolic Networks and Pathways ,Research Article ,Heparin-binding EGF-like Growth Factor ,Protein Kinase C-alpha ,Signaling in cellular processes ,Endocrine System ,Biology ,Cell Growth ,Humans ,Protein Kinase Inhibitors ,Protein kinase C ,Oncogenic Signaling ,Endocrine Physiology ,Epidermal Growth Factor ,lcsh:R ,Proteins ,Protein kinase C signaling ,Protein Structure, Tertiary ,Transmembrane Proteins ,ADAM Proteins ,lcsh:Q ,Protein Processing, Post-Translational - Abstract
Epidermal growth factor receptor (EGFR) signalling is initiated by the release of EGFR-ligands from membrane-anchored precursors, a process termed ectodomain shedding. This proteolytic event, mainly executed by A Disintegrin And Metalloproteases (ADAMs), is regulated by a number of signal transduction pathways, most notably those involving protein kinase C (PKC). However, the molecular mechanisms of PKC-dependent ectodomain shedding of EGFR-ligands, including the involvement of specific PKC isoforms and possible functional redundancy, are poorly understood. To address this issue, we employed a cell-based system of PMA-induced PKC activation coupled with shedding of heparin binding (HB)-EGF. In agreement with previous studies, we demonstrated that PMA triggers a rapid ADAM17-mediated release of HB-EGF. However, PMA-treatment also results in a protease-independent loss of cell surface HB-EGF. We identified PKCα as the key participant in the activation of ADAM17 and suggest that it acts in parallel with a pathway linking PKCδ and ERK activity. While PKCα specifically regulated PMA-induced shedding, PKCδ and ERK influenced both constitutive and inducible shedding by apparently affecting the level of HB-EGF on the cell surface. Together, these findings indicate the existence of multiple modes of regulation controlling EGFR-ligand availability and subsequent EGFR signal transduction.
- Published
- 2011
46. Anomalous inhibition of c-Met by the kinesin inhibitor aurintricarboxylic acid
- Author
-
Stéphanie Kermorgant, Peter J. Parker, Simone Radtke, Virginie Carrière, Mina Milanovic, Carine Joffre, Nick Peel, and Michael Howell
- Subjects
Cancer Research ,C-Met ,Lung Neoplasms ,Down-Regulation ,Kinesins ,Protein tyrosine phosphatase ,Tropomyosin receptor kinase C ,Receptor tyrosine kinase ,chemistry.chemical_compound ,Allosteric Regulation ,Cell Movement ,Cell Line, Tumor ,Aurintricarboxylic acid ,Humans ,Kinase activity ,Phosphorylation ,biology ,Aurintricarboxylic Acid ,Proto-Oncogene Proteins c-met ,Molecular biology ,Cell biology ,Oncology ,chemistry ,ROR1 ,biology.protein ,Tyrosine kinase ,HeLa Cells ,Signal Transduction - Abstract
c-Met [the hepatocyte growth factor (HGF) receptor] is a receptor tyrosine kinase playing a role in various biological events. Overexpression of the receptor has been observed in a number of cancers, correlating with increased metastatic tendency and poor prognosis. Additionally, activating mutations in c-Met kinase domain have been reported in a subset of familial cancers causing resistance to treatment. Receptor trafficking, relying on the integrity of the microtubule network, plays an important role in activation of downstream targets and initiation of signalling events. Aurintricarboxylic acid (ATA) is a triphenylmethane derivative that has been reported to inhibit microtubule motor proteins kinesins. Additional reported properties of this inhibitor include inhibition of protein tyrosine phosphatases, nucleases and members of the Jak family. Here we demonstrate that ATA prevents HGF-induced c-Met phosphorylation, internalisation, subsequent receptor trafficking and degradation. In addition, ATA prevented HGF-induced downstream signalling which also affected cellular function, as assayed by collective cell migration of A549 cells. Surprisingly, the inhibitory effect of ATA on HGF-induced phosphorylation and signalling in vivo was associated with an increase in basal c-Met kinase activity in vitro. It is concluded that the inhibitory effects of ATA on c-Met in vivo is an allosteric effect mediated through the kinase domain of the receptor. As the currently tested adenosine triphosphate competitive tyrosine kinase inhibitors (TKIs) may lead to tumor resistance (McDermott U, et al., Cancer Res 2010;70:1625–34), our findings suggest that novel anti-c-Met therapies could be developed in the future for cancer treatment.
- Published
- 2010
47. Manipulating signal delivery - plasma-membrane ERK activation in aPKC-dependent migration
- Author
-
Peter J. Parker, Carine Rossé, Michael Howell, and Katrina Boeckeler
- Subjects
MAPK/ERK pathway ,MAP Kinase Signaling System ,MAP Kinase Kinase 1 ,Exocyst ,Biology ,Kidney ,Focal adhesion ,Cell Movement ,Animals ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Protein kinase C ,Cells, Cultured ,Protein Kinase C ,Sirolimus ,Focal Adhesions ,Mechanism (biology) ,Cell Membrane ,SUPERFAMILY ,Cell Biology ,Cell biology ,Rats ,Enzyme Activation ,Signalling ,Membrane ,Paxillin ,Protein Binding ,Signal Transduction - Abstract
Members of the PKC superfamily have been implicated in various migratory models and in particular in spatially restricted processes. However, defining the precise local events that underlie the PKC-dependent processes is constrained by the unspecific nature of interventions. Here we address this problem in relation to atypical PKC (aPKC) action, which in conjunction with the exocyst complex controls the polarised delivery of promigratory signals. A drug-dependent recruitment approach was employed to manipulate the local recruitment of signals to the leading edge of migrating cells, under conditions where the aPKC-exocyst control is globally abrogated. We found that activation of ERK but not JNK at focal adhesions recovers the majority of the migratory loss attributed to ERK action, demonstrating a necessary role for active plasma membrane ERK in the downstream signalling of aPKC-dependent migration. The data further show that restored focal adhesion dynamics are a contributing mechanism through which localized ERK activity influences this aPKC-exocyst-dependent migration.
- Published
- 2010
48. Arkadia activates Smad3/Smad4-dependent transcription by triggering signal-induced SnoN degradation
- Author
-
Debipriya Das, Michael Howell, Laurence Levy, Caroline S. Hill, Sean Harkin, and Vasso Episkopou
- Subjects
Scaffold protein ,HECT domain ,animal structures ,Transcription, Genetic ,Ubiquitin-Protein Ligases ,SMAD ,Adenocarcinoma ,Cell Line ,Barrett Esophagus ,Mice ,Ubiquitin ,Genes, Reporter ,Transforming Growth Factor beta ,Proto-Oncogene Proteins ,Animals ,Humans ,Smad3 Protein ,RNA, Small Interfering ,Molecular Biology ,Smad4 Protein ,biology ,integumentary system ,Intracellular Signaling Peptides and Proteins ,Nuclear Proteins ,Cell Biology ,Transforming growth factor beta ,Articles ,Molecular biology ,Protein ubiquitination ,Ubiquitin ligase ,Cell biology ,Gene Expression Regulation ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Signal Transduction - Abstract
The transforming growth factor β (TGF-β) superfamily of ligands comprises TGF-βs, Activin/Nodal family members, bone morphogenetic proteins (BMPs), and growth and differentiation factors (26). These ligands signal through a heterotetrameric complex of two type II receptors and two type I receptors, both serine/threonine kinases. The ligand brings the receptors together, enabling the type II receptor to phosphorylate and activate the type I receptor. The activated type I receptor signals to the nucleus primarily through phosphorylation of receptor-regulated Smads (R-Smads) (12). Broadly speaking, TGF-β and Activin/Nodal ligands induce activation of the R-Smads, Smad2 and Smad3, while the BMP and growth and differentiation factor ligands induce activation of Smad1, -5, and -8. Activated R-Smads form homomeric complexes and heteromeric complexes with Smad4 which accumulate in the nucleus. There they are recruited to promoter elements in conjunction with other transcription factors to regulate transcription both positively and negatively. Different Smad complexes target different promoter elements. Smad3/Smad4 complexes bind directly to direct or inverted repeats of the GTCT sequence or its reverse complement, AGAC (44), such as those found in the PAI-1 promoter (6) or c-Jun promoter (41). A spliced variant of Smad2 (Smad2Δexon3) also binds as a complex with Smad4 to these same repeated GTCT or AGAC sequences (5, 42). Complexes of Smad4 with Smad1 or Smad5 also bind DNA directly and have recently been shown to recognize a GRCKNCN5GTCT consensus in cooperation with the zinc finger protein Schnurri (43). Such BMP-responsive elements (BREs) are found in the Id1 promoter (20). Full-length Smad2 cannot bind DNA directly; thus, Smad2/Smad4 complexes are recruited to DNA via other transcription factors, the best characterized being members of the FoxH1 family (3) and Mix family (13). The relatively simple Smad pathway is subject to multiple levels of regulation which allows the pathway to be fine tuned and modulated by other growth factor signaling pathways and the cell cycle (12). The pathway is also regulated by negative-feedback mechanisms which limit the duration of Smad signaling. E3 ubiquitin ligases are emerging as important negative regulators of TGF-β signaling pathways (17). Protein ubiquitination occurs in three stages utilizing E1 (ubiquitin-activating), E2 (ubiquitin-conjugating), and E3 (ubiquitin ligase) enzymes (32). E3 ubiquitin ligases are predominantly of two types: those that contain RING fingers and those that contain HECT domains. They interact specifically with the substrate, and they facilitate (RING finger E3s) or catalyze (HECT domain E3s) the transfer of ubiquitin from the E2 enzyme, respectively. The HECT domain-containing protein Smurf1 (Smad ubiquitination regulatory factor 1) was the first E3 ubiquitin ligase shown to be involved in TGF-β signaling. It binds Smad1 and Smad5 through its WW domain and a PY motif in the Smads and induces ubiquitination and degradation of these Smads (45). A close family member, Smurf2, was then shown to regulate levels of Smad1 and Smad2 (17). Smurf2 may also degrade activated R-Smads, as the association between Smurf2 and Smad2 or Smad3 is promoted by TGF-β signaling (17). Other E3 ubiquitin ligases preferentially degrade phosphorylated R-Smads, such as the multisubunit RING E3 ubiquitin ligase, Skp-1/Cul/Fbox complex which targets phosphorylated Smad3, and the HECT domain E3 ligases, Nedd4-2 and WWP1/Tiul1, which target phosphorylated Smad2. Like the R-Smads, Smad4 is regulated by E3 ligases and Smurf1/2, Nedd4-2, and WWP1/Tiul1, as well as the RING finger protein, Ectodermin/Tif1γ, have all been implicated in Smad4 degradation (8, 29). The Smurfs also have other targets in the cell. They are recruited via the inhibitory Smads, Smad6 and Smad7, to the activated TGF-β, Activin, and BMP-type I receptors and induce their degradation (9, 18). This provides a negative-feedback mechanism to terminate signaling. These E3 ubiquitin ligases also promote TGF-β signaling by degrading repressors of the pathway. The transcriptional repressors Ski and SnoN interact with activated Smad2 and Smad3 and also Smad4 and have been thought to repress transcription by disrupting formation of active heteromeric Smad complexes, recruiting transcriptional corepressor complexes, and blocking interaction of activated Smads with transcriptional activators (25). SnoN (37) and to a lesser extent, Ski (38) are ubiquitinated and degraded rapidly via the proteasome upon TGF-β stimulation. This requires Smad2 or Smad3, and lysines 440, 446, and 449 of SnoN have been shown to be required for SnoN ubiquitination (1, 36, 39). The E3 ubiquitin ligases so far implicated in this process are Smurf2, which is recruited to SnoN via Smad2 or the anaphase-promoting complex (APC), which is recruited via Smad3 (1, 36, 39). Unlike most other E3 ubiquitin ligases that modulate the TGF-β signaling pathway, Arkadia, a RING finger E3 ubiquitin ligase encoded by the gene, RNF111, was identified as a protein that enhances a subset of responses mediated by the TGF-β family member Nodal during early mouse and Xenopus embryonic development (11, 31). Subsequent work has indicated that Arkadia binds to Smad7, an inhibitory Smad, and causes its degradation. The lowering of basal levels of Smad7 in this way is thought to enhance both TGF-β and BMP signaling (19). It has recently been shown that Axin acts as a scaffold protein and cooperates with Arkadia to promote degradation of Smad7 (24). To produce a comprehensive picture of the roles of E3 ubiquitin ligases in the TGF-β signaling pathway, we undertook a small interfering RNA (siRNA) screen of 289 well-annotated human E3 ubiquitin ligases and related proteins from the RefSeq database using a HaCaT cell line containing a stably integrated Smad3/Smad4-dependent luciferase reporter, CAGA12-Luc (6). Strikingly, we found in this screen that only knockdown of Arkadia abolished TGF-β-induced transcription to the same extent as knocking down components of the pathway, like Smad3 and Smad4. Since this would not be expected from the modulatory role ascribed to Arkadia in the literature, we investigated the mechanism of Arkadia function. Our data indicate that Arkadia functions to specifically promote transcription via Smad3/Smad4 binding sites by degrading the transcriptional repressor, SnoN, in response to TGF-β signaling.
- Published
- 2007
49. Smads orchestrate specific histone modifications and chromatin remodeling to activate transcription
- Author
-
W. Lee Kraus, Sarah Ross, Michael Howell, Edwin Cheung, Thodoris G. Petrakis, and Caroline S. Hill
- Subjects
Transcriptional Activation ,Chromosomal Proteins, Non-Histone ,Response element ,E-box ,Transcription coregulator ,P300-CBP Transcription Factors ,Smad2 Protein ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Chromatin remodeling ,Article ,Histones ,Mice ,Transforming Growth Factor beta ,Animals ,Humans ,p300-CBP Transcription Factors ,Phosphorylation ,Molecular Biology ,Cells, Cultured ,Smad4 Protein ,R-SMAD ,General Immunology and Microbiology ,General transcription factor ,Models, Genetic ,General Neuroscience ,Pioneer factor ,Nuclear Proteins ,Acetylation ,Chromatin Assembly and Disassembly ,Molecular biology ,Chromatin ,Cell biology ,NIH 3T3 Cells ,biological phenomena, cell phenomena, and immunity ,HeLa Cells ,Protein Binding ,Transcription Factors - Abstract
Smads are intracellular transducers for TGF-beta superfamily ligands, but little is known about the mechanism by which complexes of receptor-phosphorylated Smad2 and Smad4 regulate transcription. Using an in vitro transcription system, we have discovered that, unlike most transcription factors that are sufficient to recruit the basal transcription machinery and therefore activate transcription on both naked DNA and chromatin templates, the Smads only activate transcription from chromatin templates. We demonstrate that Smad2-mediated transcription requires the histone acetyltransferase, p300. Smad2-recruited p300 exhibits an altered substrate specificity, specifically acetylating nucleosomal histone H3 at lysines 9 and 18, and these modifications are also detected on an endogenous Smad2-dependent promoter in a ligand-induced manner. Furthermore, we show that endogenous Smad2 interacts with the SWI/SNF ATPase, Brg1, in a TGF-beta-dependent manner, and demonstrate that Brg1 is recruited to Smad2-dependent promoters and is specifically required for TGF-beta-induced expression of endogenous Smad2 target genes. Our data indicate that the Smads define a new class of transcription factors that absolutely require chromatin to assemble the basal transcription machinery and activate transcription.
- Published
- 2006
50. Defects in Yolk Sac Vasculogenesis, Chorioallantoic Fusion, and Embryonic Axis Elongation in Mice with Targeted Disruption of Yap65
- Author
-
Michael Howell, Sharon L. Milgram, Brian N. Boone, Terry Magnuson, Jaclyn R. Stonebraker, Wanda K. O'Neal, Elizabeth M. Morin-Kensicki, Jeremy Teed, and James G. Alb
- Subjects
Embryonic Development ,Gene Expression ,Neovascularization, Physiologic ,Cell Cycle Proteins ,Biology ,Chorioallantoic Membrane ,Mice ,Vasculogenesis ,medicine ,Animals ,Yolk sac ,Molecular Biology ,Axis elongation ,Adaptor Proteins, Signal Transducing ,Yolk Sac ,Embryogenesis ,Homozygote ,Proteins ,Allantois ,YAP-Signaling Proteins ,Cell Biology ,Anatomy ,Articles ,Embryo, Mammalian ,Phosphoproteins ,Embryonic stem cell ,Mice, Mutant Strains ,Cell biology ,Neuroepithelial cell ,Chorioallantoic membrane ,medicine.anatomical_structure ,embryonic structures ,Gene Targeting ,Mutation ,Genes, Lethal ,Acyltransferases ,Transcription Factors - Abstract
YAP is a multifunctional adapter protein and transcriptional coactivator with several binding partners well described in vitro and in cell culture. To explore in vivo requirements for YAP, we generated mice carrying a targeted disruption of the Yap gene. Homozygosity for the Yap(tm1Smil) allele (Yap-/-) caused developmental arrest around E8.5. Phenotypic characterization revealed a requirement for YAP in yolk sac vasculogenesis. Yolk sac endothelial and erythrocyte precursors were specified as shown by histology, PECAM1 immunostaining, and alpha globin expression. Nonetheless, development of an organized yolk sac vascular plexus failed in Yap-/- embryos. In striking contrast, vasculogenesis proceeded in both the allantois and the embryo proper. Mutant embryos showed patterned gene expression domains along the anteroposterior neuraxis, midline, and streak/tailbud. Despite this evidence of proper patterning and tissue specification, Yap-/- embryos showed developmental perturbations that included a notably shortened body axis, convoluted anterior neuroepithelium, caudal dysgenesis, and failure of chorioallantoic fusion. These results reveal a vital requirement for YAP in the developmental processes of yolk sac vasculogenesis, chorioallantoic attachment, and embryonic axis elongation.
- Published
- 2006
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.