Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Development ; 147(21)2020 11 12.
Article in English | MEDLINE | ID: mdl-32994166

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bone and Bones/embryology , Bone and Bones/metabolism , Cell Cycle Proteins/metabolism , Morphogenesis , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/metabolism , Animals , Bone and Bones/abnormalities , Bone and Bones/pathology , Cartilage/pathology , Cell Nucleus/metabolism , Cell Proliferation , Chondrocytes/metabolism , Chondrocytes/pathology , Cleft Palate/pathology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Gene Expression Regulation, Developmental , Growth Plate/pathology , Hippo Signaling Pathway , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis/genetics , Signal Transduction , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins
2.
Nucleic Acids Res ; 47(6): 2906-2921, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30590722

ABSTRACT

The Topo2a-dependent arrest is associated with faithful segregation of sister chromatids and has been identified as dysfunctional in numerous tumour cell lines. This genome-protecting pathway is poorly understood and its characterization is of significant interest, potentially offering interventional opportunities in relation to synthetic lethal behaviours in arrest-defective tumours. Using the catalytic Topo2a inhibitor ICRF193, we have performed a genome-wide siRNA screen in arrest-competent, non-transformed cells, to identify genes essential for this arrest mechanism. In addition, we have counter-screened several DNA-damaging agents and demonstrate that the Topo2a-dependent arrest is genetically distinct from DNA damage checkpoints. We identify the components of the SMC5/6 complex, including the activity of the E3 SUMO ligase NSE2, as non-redundant players that control the timing of the Topo2a-dependent arrest in G2. We have independently verified the NSE2 requirement in fibroblasts from patients with germline mutations that cause severely reduced levels of NSE2. Through imaging Topo2a-dependent G2 arrested cells, an increased interaction between Topo2a and NSE2 is observed at PML bodies, which are known SUMOylation hotspots. We demonstrate that Topo2a is SUMOylated in an ICRF193-dependent manner by NSE2 at a novel non-canonical site (K1520) and that K1520 sumoylation is required for chromosome segregation but not the G2 arrest.


Subject(s)
DNA Topoisomerases, Type II/genetics , G2 Phase Cell Cycle Checkpoints/genetics , Ligases/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Sumoylation/genetics , Cell Cycle Proteins/genetics , Cell Line , Chromosomal Proteins, Non-Histone/genetics , DNA Damage/drug effects , Diketopiperazines , Fibroblasts/drug effects , Genome, Human/genetics , Germ-Line Mutation/genetics , Humans , Multiprotein Complexes/genetics , Piperazines/pharmacology , Poly-ADP-Ribose Binding Proteins/antagonists & inhibitors , RNA Interference , Ubiquitin-Protein Ligases/genetics
3.
EMBO J ; 31(8): 1931-46, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22354037

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.


Subject(s)
Amino Acids/metabolism , Autophagy , Carrier Proteins/metabolism , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Carrier Proteins/antagonists & inhibitors , Cell Line , Gene Silencing , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Membrane Proteins/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Phagosomes/metabolism , RNA, Small Interfering/metabolism , RNA-Binding Proteins , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Staining and Labeling
4.
bioRxiv ; 2024 Mar 16.
Article in English | MEDLINE | ID: mdl-38559242

ABSTRACT

Immunomodulatory imide drugs (IMiDs) including thalidomide, lenalidomide, and pomalidomide, can be used to induce degradation of a protein of interest that is fused to a short zinc finger (ZF) degron motif. These IMiDs, however, also induce degradation of endogenous neosubstrates, including IKZF1 and IKZF3. To improve degradation selectivity, we took a bump-and-hole approach to design and screen bumped IMiD analogs against 8380 ZF mutants. This yielded a bumped IMiD analog that induces efficient degradation of a mutant ZF degron, while not affecting other cellular proteins, including IKZF1 and IKZF3. In proof-of-concept studies, this system was applied to induce efficient degradation of TRIM28, a disease-relevant protein with no known small molecule binders. We anticipate that this system will make a valuable addition to the current arsenal of degron systems for use in target validation.

5.
J Pathol ; 227(2): 146-56, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22362593

ABSTRACT

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 overexpressed 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 eight ccRCC lines tested and was the seventh most overexpressed gene in a meta-analysis of five 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. Intra-tumoural 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.


Subject(s)
Carcinoma, Renal Cell/genetics , Glycolysis/genetics , Kidney Neoplasms/genetics , Monocarboxylic Acid Transporters/genetics , Muscle Proteins/genetics , RNA Interference , Apoptosis , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , Cell Survival , Disease-Free Survival , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Hydrogen-Ion Concentration , Kaplan-Meier Estimate , Kidney Neoplasms/metabolism , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Lactic Acid/metabolism , Phenotype , Prognosis , RNA, Messenger/metabolism , Time Factors , Transfection
6.
Thromb Haemost ; 102(2): 287-301, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19652879

ABSTRACT

Factor XI (FXI) functions in blood coagulation. FXI is composed of four apple (Ap) domains and a serine protease (SP) domain. Deficiency of FXI leads to an injury-related bleeding disorder, which is remarkable for the lack of correlation between bleeding symptoms and FXI coagulant activity (FXI:C). The number of mutations previously reported in our interactive web database (http://www.FactorXI.org) is now significantly increased to 183 through our new patient studies and from literature surveys. Eight novel missense mutations give a total of 120 throughout the FXI gene (F11). The most abundant defects in FXI are revealed to be those from low-protein plasma levels (Type I: CRM-) that originate from protein misfolding, rather than from functional defects (Type II: CRM+). A total of 70 Ap missense mutations were analysed using a consensus Ap domain structure generated from the FXI dimer crystal structure. This showed that all parts of the Ap domain were affected. The 47 SP missense mutations were also distributed throughout the SP domain structure. The periphery of the Ap beta-sheet structure is sensitive to structural perturbation caused by residue changes throughout the Ap domain, yet this beta-sheet is crucial for FXI dimer formation. Residues located at the Ap4:Ap4 interface in the dimer are much less directly involved. We conclude that the abundance of Type I defects in FXI results from the sensitivity of the Ap domain folding to residue changes within this, and discuss how structural knowledge of the mutations improves our understanding of FXI deficiencies.


Subject(s)
Factor XI Deficiency/genetics , Factor XI/chemistry , Factor XI/genetics , Mutation, Missense , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Crystallography, X-Ray , DNA/genetics , Databases, Genetic , Dimerization , Factor XI Deficiency/blood , Genes, Dominant , Humans , Models, Molecular , Molecular Sequence Data , Phenotype , Polymorphism, Single Nucleotide , Protein Folding , Protein Structure, Quaternary , Protein Structure, Tertiary , Sequence Homology, Amino Acid
7.
Cancer Res ; 79(8): 1884-1898, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30765601

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is driven by metabolic changes in pancreatic cells caused by oncogenic mutations and dysregulation of p53. PDAC cell lines and PDAC-derived xenografts grow as a result of altered metabolic pathways, changes in stroma, and autophagy. Selective targeting and inhibition of one of these may open avenues for the development of new therapeutic strategies. In this study, we performed a genome-wide siRNA screen in a PDAC cell line using endogenous autophagy as a readout and identified several regulators of autophagy that were required for autophagy-dependent PDAC cell survival. Validation of two promising candidates, MPP7 (MAGUK p55 subfamily member 7, a scaffolding protein involved in cell-cell contacts) and MDH1 (cytosolic Malate dehydrogenase 1), revealed their role in early stages of autophagy during autophagosome formation. MPP7 was involved in the activation of YAP1 (a transcriptional coactivator in the Hippo pathway), which in turn promoted autophagy, whereas MDH1 was required for maintenance of the levels of the essential autophagy initiator serine-threonine kinase ULK1, and increased in the activity upon induction of autophagy. Our results provide a possible explanation for how autophagy is regulated by MPP7 and MDH1, which adds to our understanding of autophagy regulation in PDAC. SIGNIFICANCE: This study identifies and characterizes MPP7 and MDH1 as novel regulators of autophagy, which is thought to be responsible for pancreatic cancer cell survival.


Subject(s)
Autophagy , Carcinoma, Pancreatic Ductal/pathology , Gene Expression Regulation, Neoplastic , Malate Dehydrogenase/metabolism , Membrane Proteins/metabolism , Pancreatic Neoplasms/pathology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Proliferation , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Malate Dehydrogenase/antagonists & inhibitors , Malate Dehydrogenase/genetics , Membrane Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , RNA, Small Interfering/genetics , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured , YAP-Signaling Proteins
8.
Hum Mutat ; 29(3): 333-44, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18058827

ABSTRACT

Central repositories of mutations that combine structural, sequence, and phenotypic information in related proteins will facilitate the diagnosis and molecular understanding of diseases associated with them. Coagulation involves the sequential activation of serine proteases and regulators in order to yield stable blood clots while maintaining hemostasis. Five coagulation serine proteases-factor VII (F7), factor IX (F9), factor X (F10), protein C (PROC), and thrombin (F2)-exhibit high sequence similarities and all require vitamin K. All five of these were incorporated into an interactive database of mutations named CoagMDB (http://www.coagMDB.org; last accessed: 9 August 2007). The large number of mutations involved (especially for factor IX) and the increasing problem of out-of-date databases required the development of new database management tools. A text mining tool automatically scans full-length references to identify and extract mutations. High recall rates between 96 and 99% and precision rates of 87 to 93% were achieved. Text mining significantly reduces the time and expertise required to maintain the databases and offers a solution to the problem of locus-specific database management and upkeep. A total of 875 mutations were extracted from 1,279 literature sources. Of these, 116 correspond to Gla domains, 86 to the N-terminal EGF domain, 73 to the C-terminal EGF domain, and 477 to the serine protease domain. The combination of text mining and consensus domain structures enables mutations to be correlated with experimentally-measurable phenotypes based on either low protein levels (Type I) or reduced functional activities (Type II), respectively. A tendency for the conservation of phenotype with structural location was identified.


Subject(s)
Blood Coagulation Factors/genetics , Databases, Genetic , Mutation, Missense , Serine Endopeptidases/genetics , Algorithms , Amino Acid Sequence , Amino Acid Substitution , Blood Coagulation Factors/chemistry , Consensus Sequence , Conserved Sequence/genetics , Factor IX/chemistry , Factor IX/genetics , Factor VII/chemistry , Factor VII/genetics , Factor X/chemistry , Factor X/genetics , Humans , Models, Genetic , Models, Molecular , Molecular Sequence Data , Natural Language Processing , Protein C/chemistry , Protein C/genetics , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Serine Endopeptidases/chemistry , Thrombin/chemistry , Thrombin/genetics , Vitamin K/metabolism
9.
BMC Struct Biol ; 8: 19, 2008 Mar 25.
Article in English | MEDLINE | ID: mdl-18366741

ABSTRACT

BACKGROUND: Phosphorylation is a central feature in many biological processes. Structural analyses have identified the importance of charge-charge interactions, for example mediating phosphorylation-driven allosteric change and protein binding to phosphopeptides. Here, we examine computationally the prevalence of charge stabilisation around phosphorylated sites in the structural database, through comparison with locations that are not phosphorylated in the same structures. RESULTS: A significant fraction of phosphorylated sites appear to be electrostatically stabilised, largely through interaction with sidechains. Some examples of stabilisation across a subunit interface are evident from calculations with biological units. When considering the immediately surrounding environment, in many cases favourable interactions are only apparent after conformational change that accompanies phosphorylation. A simple calculation of potential interactions at longer-range, applied to non-phosphorylated structures, recovers the separation exhibited by phosphorylated structures. In a study of sites in the Phospho.ELM dataset, for which structural annotation is provided by non-phosphorylated proteins, there is little separation of the known phospho-acceptor sites relative to background, even using the wider interaction radius. However, there are differences in the distributions of patch polarity for acceptor and background sites in the Phospho.ELM dataset. CONCLUSION: In this study, an easy to implement procedure is developed that could contribute to the identification of phospho-acceptor sites associated with charge-charge interactions and conformational change. Since the method gives information about potential anchoring interactions subsequent to phosphorylation, it could be combined with simulations that probe conformational change. Our analysis of the Phospho.ELM dataset also shows evidence for mediation of phosphorylation effects through (i) conformational change associated with making a solvent inaccessible phospho-acceptor site accessible, and (ii) modulation of protein-protein interactions.


Subject(s)
Proteins/chemistry , Serine/chemistry , Threonine/chemistry , Tyrosine/chemistry , Binding Sites , Databases, Protein , Models, Molecular , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Proteins/metabolism , Serine/metabolism , Threonine/metabolism , Tyrosine/metabolism
10.
Am J Kidney Dis ; 51(3): 487-90, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18295065

ABSTRACT

Hemolytic uremic syndrome (HUS) takes 2 forms: diarrheal HUS and nondiarrheal HUS. As its name suggests, diarrheal HUS classically follows an enteric infection. The classic infective organism is the Escherichia coli O157 serotype, although other bacteria, including Shigella species, can produce the verocytotoxin required to cause HUS. The usual clinical course is an episode of bloody diarrhea followed by thrombotic microangiopathy and acute renal failure. Supportive treatment sees recovery of renal function in the vast majority of patients. Most cases occur in children, but all age groups can be affected. Conversely, nondiarrheal HUS may have one of a number of predisposing factors, including drugs, irradiation, and hypertension. It also is well established that mutations in the genes encoding the complement regulator proteins factor H, factor I, and membrane cofactor protein predispose to nondiarrheal HUS. In patients with nondiarrheal HUS, recovery of renal function is much less common. Here, we present a case of HUS after a diarrheal illness in which the patient did not recover renal function in the long term. A novel mutation in exon 23 of the factor H gene was discovered. This is clinically important. If this patient underwent transplantation, he would be expected to have an 80% risk of graft loss at 2 years because of recurrent HUS. We recommend consideration of complement gene mutations in any patient with HUS after a diarrheal episode in which there are unusual features.


Subject(s)
Complement Factor H/genetics , Hemolytic-Uremic Syndrome/genetics , Mutation, Missense , Adult , Base Sequence , Creatinine/blood , Diarrhea/complications , Disease Progression , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/diagnosis , Humans , Male , Mutation, Missense/genetics , Sequence Analysis, DNA
11.
Cell Rep ; 25(7): 1841-1855.e5, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30428352

ABSTRACT

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 ß (TGF-ß) 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-ß 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.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Activins/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Line , Down-Regulation , Epithelial-Mesenchymal Transition , Genome, Human , Humans , Lysosomes/metabolism , Mice , Multivesicular Bodies/metabolism , Neoplasms/pathology , Phosphorylation , Prognosis , Protein Transport , Proteolysis , Smad2 Protein/metabolism , Survival Analysis , Ubiquitin-Protein Ligases/metabolism , Up-Regulation
12.
Hum Mutat ; 28(3): 222-34, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17089378

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) is a disease of hemolytic anemia, thrombocytopenia, and renal failure associated with defective alternative pathway (AP) complement control. Previously, we presented a database (www.FH-HUS.org) focusing on aHUS mutations in the Factor H gene (CFH). Here, new aHUS mutations are reported for the complement regulatory proteins Factor H (FH), Factor I (FI), and membrane cofactor protein (MCP). Additional mutations or polymorphisms within CFH have been associated with membranoproliferative glomerulonephritis (MPGN) and age-related macular degeneration (AMD). Accordingly, the database now includes substitutions that predispose to aHUS, MPGN, and AMD. For this, structural models for the domains in MCP and FI were developed using homology modeling. With this new database, patients with mutations in more than one gene can be displayed and interpreted in a coherent manner. The database also includes SNP polymorphisms in CFH, MCP, and IF. There are now a total of 167 genetic alterations, including 100 in CFH, 43 in MCP, and 24 in IF. The mutations characterize clinical outcomes that vary from several AMD-associated polymorphisms to those associated with aHUS, MPGN, or FI deficiency. A consensus short complement regulator (SCR) domain structure facilitated the interpretations of aHUS mutations. Specific locations within this consensus domain often correlate with the occurrence of clinical phenotypes. The AMD Tyr402His polymorphism is structurally located at a hotspot for several aHUS mutations. The database emphasizes the causative role of the alternative pathway of complement in disease and provides a repository of knowledge to assist future diagnosis and novel therapeutic approaches.


Subject(s)
Complement Factor H/genetics , Databases, Genetic , Fibrinogen/genetics , Hemolytic-Uremic Syndrome/genetics , Membrane Cofactor Protein/genetics , Amino Acid Sequence , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Protein Structure, Tertiary/genetics , Sequence Homology, Amino Acid
13.
Hum Mutat ; 27(1): 21-30, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16281287

ABSTRACT

Factor H (FH) is a central complement regulator comprised of 20 short complement repeat (SCR) domains. Nucleotide changes within this gene (CFH) have been observed in patients with hemolytic uremic syndrome (HUS), and also membranoproliferative glomerulonephritis and age-related macular degeneration. All parts of FH are affected, but many mutations are clustered in the C-terminal part of FH. Up to now, structural analyses of HUS have been based on SCR-20, a domain that is involved in FH interactions with C3b, heparin, and endothelial cells. In order to identify the structural and functional consequence of HUS mutations, further disease-associated mutations were analyzed in terms of homology and nuclear magnetic resonance (NMR) models for factor H SCR domains. An interactive web database of 54 human HUS-associated mutations and others was created from the literature (www.FH-HUS.org). This has comprehensive search and analysis tools, integrating phenotypic and genetic data with structural analysis. Each mutation can be highlighted on the SCR structure together with the patient FH and C3 levels where available. Two new insights were obtained from our collection of data. First, phenotypic data on FH clarify our previously-proposed classification of Type I and Type II disorders that both lead to HUS, where Type I affects FH secretion and folding, and Type II leads to expressed protein in plasma that is functionally defective. Second, the new mutations show more clearly that SCR domains from SCR-16 to SCR-19 are important for the ligand binding activities of FH as well as SCR-20. This FH web database will facilitate the interpretation of new mutations and polymorphisms when these are identified in patients, and it will clarify the functional role of FH.


Subject(s)
Complement Factor H/genetics , Databases, Genetic , Hemolytic-Uremic Syndrome/genetics , Internet , Mutation/genetics , Amino Acid Sequence , Complement Factor H/chemistry , Genetic Predisposition to Disease , Humans , Models, Molecular , Molecular Sequence Data , Structural Homology, Protein
14.
Cell Rep ; 15(7): 1597-1610, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27184836

ABSTRACT

In order to facilitate the identification of factors and pathways in the cellular response to UV-induced DNA damage, several descriptive proteomic screens and a functional genomics screen were performed in parallel. Numerous factors could be identified with high confidence when the screen results were superimposed and interpreted together, incorporating biological knowledge. A searchable database, bioLOGIC, which provides access to relevant information about a protein or process of interest, was established to host the results and facilitate data mining. Besides uncovering roles in the DNA damage response for numerous proteins and complexes, including Integrator, Cohesin, PHF3, ASC-1, SCAF4, SCAF8, and SCAF11, we uncovered a role for the poorly studied, melanoma-associated serine/threonine kinase 19 (STK19). Besides effectively uncovering relevant factors, the multiomic approach also provides a systems-wide overview of the diverse cellular processes connected to the transcription-related DNA damage response.


Subject(s)
DNA Damage/radiation effects , Proteomics , Ultraviolet Rays , Chromatin/metabolism , Databases, Factual , HEK293 Cells , Humans , Internet , Leupeptins/pharmacology , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/radiation effects , Nuclear Proteins/metabolism , Phosphorylation/radiation effects , Protein Serine-Threonine Kinases/metabolism , Proteome/drug effects , Proteome/radiation effects , RNA Polymerase II/metabolism , RNA, Small Interfering/metabolism , Transcription, Genetic/radiation effects , Ubiquitination/radiation effects , User-Computer Interface
15.
Hum Mutat ; 26(3): 192-8, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16086308

ABSTRACT

Factor XI (FXI) is the zymogen of a serine protease enzyme in the intrinsic pathway of blood coagulation and is an important factor in the creation of a stable fibrin clot. Deficiency of FXI leads to an injury-related bleeding disorder and is remarkable for the lack of correlation between bleeding symptoms and FXI coagulant activity (FXI:C). The FXI protein is composed of five domains: four tandem repeat domains of approximately 80 residues known as Apple (Ap) domains, and the catalytic serine protease (Sp) domain. A total of 65 mutations throughout the FXI gene (F11) have been reported in FXI deficient patients. An interactive web database of these mutations has been created (www.FactorXI.org) that integrates the phenotypic data with genetic data and structural homology models for the five FXI domains. The database provides a central repository for all reported genetic alterations within F11. With the use of recently developed visualization tools, each mutation can be highlighted on the structural models of the FXI domains together with an appropriate survey of patient data, such as FXI:C levels and FXI antigen levels. The database also enables new F11 mutations to be interpreted. The interactive design of this database will lead to a more comprehensive comparative understanding of the genetic factors that influence bleeding risk.


Subject(s)
Databases, Genetic , Factor XI Deficiency/genetics , Factor XI/genetics , Mutation , DNA Mutational Analysis , Disulfides/chemistry , Humans , Models, Genetic , Models, Molecular , Molecular Conformation , Phenotype , Protein Structure, Tertiary , Risk
16.
Cancer Cell ; 27(1): 57-71, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25584894

ABSTRACT

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.


Subject(s)
Acetate-CoA Ligase/genetics , Acetate-CoA Ligase/metabolism , Fatty Acids/metabolism , Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation , Disease Progression , Gene Dosage , Gene Expression Regulation, Neoplastic , Humans , Hypoxia , MCF-7 Cells , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms/genetics , Neoplasms/metabolism , Stress, Physiological
17.
Cancer Discov ; 4(5): 606-19, 2014 May.
Article in English | MEDLINE | ID: mdl-24535670

ABSTRACT

Activating mutations in the EGF receptor (EGFR) are associated with clinical responsiveness to EGFR tyrosine kinase inhibitors (TKI), such as erlotinib and gefitinib. However, resistance eventually arises, often due to a second EGFR mutation, most commonly T790M. Through a genome-wide siRNA screen in a human lung cancer cell line and analyses of murine mutant EGFR-driven lung adenocarcinomas, we found that erlotinib resistance was associated with reduced expression of neurofibromin, the RAS GTPase-activating protein encoded by the NF1 gene. Erlotinib failed to fully inhibit RAS-ERK signaling when neurofibromin levels were reduced. Treatment of neurofibromin-deficient lung cancers with a MAP-ERK kinase (MEK) inhibitor restored sensitivity to erlotinib. Low levels of NF1 expression were associated with primary and acquired resistance of lung adenocarcinomas to EGFR TKIs in patients. These findings identify a subgroup of patients with EGFR-mutant lung adenocarcinoma who might benefit from combination therapy with EGFR and MEK inhibitors.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm , Erlotinib Hydrochloride/administration & dosage , Lung Neoplasms/drug therapy , Neurofibromin 1/genetics , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Erlotinib Hydrochloride/therapeutic use , Humans , Lung Neoplasms/genetics , MAP Kinase Signaling System , Mice , Neoplasms, Experimental , Neurofibromin 1/metabolism , Pyridones/therapeutic use , Pyrimidinones/therapeutic use
18.
Database (Oxford) ; 2013: bat072, 2013.
Article in English | MEDLINE | ID: mdl-24122843

ABSTRACT

High-throughput screening (HTS) uses technologies such as RNA interference to generate loss-of-function phenotypes on a genomic scale. As these technologies become more popular, many research institutes have established core facilities of expertise to deal with the challenges of large-scale HTS experiments. As the efforts of core facility screening projects come to fruition, focus has shifted towards managing the results of these experiments and making them available in a useful format that can be further mined for phenotypic discovery. The HTS-DB database provides a public view of data from screening projects undertaken by the HTS core facility at the CRUK London Research Institute. All projects and screens are described with comprehensive assay protocols, and datasets are provided with complete descriptions of analysis techniques. This format allows users to browse and search data from large-scale studies in an informative and intuitive way. It also provides a repository for additional measurements obtained from screens that were not the focus of the project, such as cell viability, and groups these data so that it can provide a gene-centric summary across several different cell lines and conditions. All datasets from our screens that can be made available can be viewed interactively and mined for further hit lists. We believe that in this format, the database provides researchers with rapid access to results of large-scale experiments that might facilitate their understanding of genes/compounds identified in their own research. DATABASE URL: http://hts.cancerresearchuk.org/db/public.


Subject(s)
Databases, Genetic , Genomics , High-Throughput Screening Assays/methods , Publishing , RNA, Small Interfering/metabolism , Search Engine , Cell Line , Cell Survival/genetics , Gene Regulatory Networks/genetics , Humans , Internet , Meta-Analysis as Topic
19.
Nat Cell Biol ; 15(1): 61-71, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23263283

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 characterize 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 Ser 413. As Sik kinases have been implicated in nutrient sensing, this suggests a link between the Hpo pathway and systemic growth control.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , 14-3-3 Proteins/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Nuclear Proteins/metabolism , Organ Size , Phosphorylation , Protein Binding , Protein Interaction Mapping , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/genetics , RNA Interference , Trans-Activators/metabolism , Wings, Animal/enzymology , Wings, Animal/growth & development , YAP-Signaling Proteins
20.
Semin Thromb Hemost ; 32(2): 160-8, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16575691

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) mutations have been reported in the complement regulatory proteins factor H, factor I, and membrane cofactor protein (MCP). Mutations within factor H are also associated with membranoproliferative glomerulonephritis and age-related macular degeneration. The increasing amount of information on aHUS requires organization if it is to be usable. Accordingly, an interactive factor H aHUS Web database has been developed (http://www.fh-hus.org) that integrates genotypic, phenotypic, and structural information for mutations within human factor H. This provides a valuable tool for the interpretation of previously reported aHUS mutations, and provides prediction and analysis tools for new mutations. It will be extended to include mutations in factor I and MCP. Here, we describe how to use this Web database as a research tool, and indicate possible future directions depending on feedback from the clinical community.


Subject(s)
Complement Factor H/genetics , Databases, Factual , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/genetics , Humans , Internet , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL