Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters

Publication year range
1.
Cell ; 178(4): 807-819.e21, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398338

ABSTRACT

The NRF2 transcription factor controls a cell stress program that is implicated in cancer and there is great interest in targeting NRF2 for therapy. We show that NRF2 activity depends on Fructosamine-3-kinase (FN3K)-a kinase that triggers protein de-glycation. In its absence, NRF2 is extensively glycated, unstable, and defective at binding to small MAF proteins and transcriptional activation. Moreover, the development of hepatocellular carcinoma triggered by MYC and Keap1 inactivation depends on FN3K in vivo. N-acetyl cysteine treatment partially rescues the effects of FN3K loss on NRF2 driven tumor phenotypes indicating a key role for NRF2-mediated redox balance. Mass spectrometry reveals that other proteins undergo FN3K-sensitive glycation, including translation factors, heat shock proteins, and histones. How glycation affects their functions remains to be defined. In summary, our study reveals a surprising role for the glycation of cellular proteins and implicates FN3K as targetable modulator of NRF2 activity in cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , NF-E2-Related Factor 2/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Female , Gene Knockdown Techniques , Glucose/metabolism , Glycosylation , HEK293 Cells , Hep G2 Cells , Heterografts , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Phosphotransferases (Alcohol Group Acceptor)/genetics , Proto-Oncogene Proteins c-myc/metabolism , Transduction, Genetic
2.
Nature ; 615(7951): 339-348, 2023 03.
Article in English | MEDLINE | ID: mdl-36859550

ABSTRACT

Trimethylation of histone H3 lysine 4 (H3K4me3) is associated with transcriptional start sites and has been proposed to regulate transcription initiation1,2. However, redundant functions of the H3K4 SET1/COMPASS methyltransferase complexes complicate the elucidation of the specific role of H3K4me3 in transcriptional regulation3,4. Here, using mouse embryonic stem cells as a model system, we show that acute ablation of shared subunits of the SET1/COMPASS complexes leads to a complete loss of all H3K4 methylation. Turnover of H3K4me3 occurs more rapidly than that of H3K4me1 and H3K4me2 and is dependent on KDM5 demethylases. Notably, acute loss of H3K4me3 does not have detectable effects on transcriptional initiation but leads to a widespread decrease in transcriptional output, an increase in RNA polymerase II (RNAPII) pausing and slower elongation. We show that H3K4me3 is required for the recruitment of the integrator complex subunit 11 (INTS11), which is essential for the eviction of paused RNAPII and transcriptional elongation. Thus, our study demonstrates a distinct role for H3K4me3 in transcriptional pause-release and elongation rather than transcriptional initiation.


Subject(s)
Histones , Mouse Embryonic Stem Cells , Promoter Regions, Genetic , RNA Polymerase II , Transcription Elongation, Genetic , Transcription Termination, Genetic , Animals , Mice , Gene Expression Regulation , Histone Demethylases/metabolism , Histones/chemistry , Histones/metabolism , Methylation , Mouse Embryonic Stem Cells/metabolism , Promoter Regions, Genetic/genetics , RNA Polymerase II/metabolism
3.
Mol Cell ; 81(8): 1749-1765.e8, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33657400

ABSTRACT

Acetylation of lysine 16 on histone H4 (H4K16ac) is catalyzed by histone acetyltransferase KAT8 and can prevent chromatin compaction in vitro. Although extensively studied in Drosophila, the functions of H4K16ac and two KAT8-containing protein complexes (NSL and MSL) are not well understood in mammals. Here, we demonstrate a surprising complex-dependent activity of KAT8: it catalyzes H4K5ac and H4K8ac as part of the NSL complex, whereas it catalyzes the bulk of H4K16ac as part of the MSL complex. Furthermore, we show that MSL complex proteins and H4K16ac are not required for cell proliferation and chromatin accessibility, whereas the NSL complex is essential for cell survival, as it stimulates transcription initiation at the promoters of housekeeping genes. In summary, we show that KAT8 switches catalytic activity and function depending on its associated proteins and that, when in the NSL complex, it catalyzes H4K5ac and H4K8ac required for the expression of essential genes.


Subject(s)
Histone Acetyltransferases/genetics , Homeostasis/genetics , Transcription, Genetic/genetics , Acetylation , Animals , Cell Line , Cell Line, Tumor , Cell Nucleus/genetics , Cell Proliferation/genetics , Chromatin/genetics , HEK293 Cells , HeLa Cells , Histones/genetics , Humans , K562 Cells , Lysine/genetics , Male , Mice , Promoter Regions, Genetic/genetics , THP-1 Cells
4.
Nature ; 604(7905): 354-361, 2022 04.
Article in English | MEDLINE | ID: mdl-35355015

ABSTRACT

Oncogenic alterations to DNA are not transforming in all cellular contexts1,2. This may be due to pre-existing transcriptional programmes in the cell of origin. Here we define anatomic position as a major determinant of why cells respond to specific oncogenes. Cutaneous melanoma arises throughout the body, whereas the acral subtype arises on the palms of the hands, soles of the feet or under the nails3. We sequenced the DNA of cutaneous and acral melanomas from a large cohort of human patients and found a specific enrichment for BRAF mutations in cutaneous melanoma and enrichment for CRKL amplifications in acral melanoma. We modelled these changes in transgenic zebrafish models and found that CRKL-driven tumours formed predominantly in the fins of the fish. The fins are the evolutionary precursors to tetrapod limbs, indicating that melanocytes in these acral locations may be uniquely susceptible to CRKL. RNA profiling of these fin and limb melanocytes, when compared with body melanocytes, revealed a positional identity gene programme typified by posterior HOX13 genes. This positional gene programme synergized with CRKL to amplify insulin-like growth factor (IGF) signalling and drive tumours at acral sites. Abrogation of this CRKL-driven programme eliminated the anatomic specificity of acral melanoma. These data suggest that the anatomic position of the cell of origin endows it with a unique transcriptional state that makes it susceptible to only certain oncogenic insults.


Subject(s)
Melanoma , Skin Neoplasms , Animals , Animals, Genetically Modified , Carcinogenesis/genetics , Foot , Hand , Humans , Melanoma/pathology , Nails , Oncogenes/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transcription, Genetic , Zebrafish/genetics , Melanoma, Cutaneous Malignant
5.
EMBO J ; 42(24): e114221, 2023 Dec 11.
Article in English | MEDLINE | ID: mdl-37987160

ABSTRACT

Efficient treatment of acute myeloid leukemia (AML) patients remains a challenge despite recent therapeutic advances. Here, using a CRISPRi screen targeting chromatin factors, we identified the nucleosome-remodeling factor (NURF) subunit BPTF as an essential regulator of AML cell survival. We demonstrate that BPTF forms an alternative NURF chromatin remodeling complex with SMARCA5 and BAP18, which regulates the accessibility of a large set of insulator regions in leukemic cells. This ensures efficient CTCF binding and boundary formation between topologically associated domains that is essential for maintaining the leukemic transcriptional programs. We also demonstrate that the well-studied PHD2-BROMO chromatin reader domains of BPTF, while contributing to complex recruitment to chromatin, are dispensable for leukemic cell growth. Taken together, our results uncover how the alternative NURF complex contributes to leukemia and provide a rationale for its targeting in AML.


Subject(s)
Drosophila Proteins , Leukemia, Myeloid, Acute , Humans , Chromatin/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Leukemia, Myeloid, Acute/genetics , Drosophila Proteins/metabolism , Chromatin Assembly and Disassembly
6.
Nature ; 578(7793): E11, 2020 02.
Article in English | MEDLINE | ID: mdl-31937917

ABSTRACT

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

7.
Nature ; 577(7791): 566-571, 2020 01.
Article in English | MEDLINE | ID: mdl-31915377

ABSTRACT

Epithelial-to-mesenchymal transitions (EMTs) are phenotypic plasticity processes that confer migratory and invasive properties to epithelial cells during development, wound-healing, fibrosis and cancer1-4. EMTs are driven by SNAIL, ZEB and TWIST transcription factors5,6 together with microRNAs that balance this regulatory network7,8. Transforming growth factor ß (TGF-ß) is a potent inducer of developmental and fibrogenic EMTs4,9,10. Aberrant TGF-ß signalling and EMT are implicated in the pathogenesis of renal fibrosis, alcoholic liver disease, non-alcoholic steatohepatitis, pulmonary fibrosis and cancer4,11. TGF-ß depends on RAS and mitogen-activated protein kinase (MAPK) pathway inputs for the induction of EMTs12-19. Here we show how these signals coordinately trigger EMTs and integrate them with broader pathophysiological processes. We identify RAS-responsive element binding protein 1 (RREB1), a RAS transcriptional effector20,21, as a key partner of TGF-ß-activated SMAD transcription factors in EMT. MAPK-activated RREB1 recruits TGF-ß-activated SMAD factors to SNAIL. Context-dependent chromatin accessibility dictates the ability of RREB1 and SMAD to activate additional genes that determine the nature of the resulting EMT. In carcinoma cells, TGF-ß-SMAD and RREB1 directly drive expression of SNAIL and fibrogenic factors stimulating myofibroblasts, promoting intratumoral fibrosis and supporting tumour growth. In mouse epiblast progenitors, Nodal-SMAD and RREB1 combine to induce expression of SNAIL and mesendoderm-differentiation genes that drive gastrulation. Thus, RREB1 provides a molecular link between RAS and TGF-ß pathways for coordinated induction of developmental and fibrogenic EMTs. These insights increase our understanding of the regulation of epithelial plasticity and its pathophysiological consequences in development, fibrosis and cancer.


Subject(s)
DNA-Binding Proteins/metabolism , Epithelial-Mesenchymal Transition , Fibrosis/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , ras Proteins/metabolism , Animals , Cell Line, Tumor , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Female , Fibrosis/pathology , Gastrulation , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Neoplasms/enzymology , Organoids/metabolism , Organoids/pathology , Smad Proteins/metabolism , Snail Family Transcription Factors/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Transforming Growth Factor beta/pharmacology
8.
Nature ; 568(7750): 112-116, 2019 04.
Article in English | MEDLINE | ID: mdl-30918399

ABSTRACT

Chimeric antigen receptors (CARs) are synthetic antigen receptors that reprogram T cell specificity, function and persistence1. Patient-derived CAR T cells have demonstrated remarkable efficacy against a range of B-cell malignancies1-3, and the results of early clinical trials suggest activity in multiple myeloma4. Despite high complete response rates, relapses occur in a large fraction of patients; some of these are antigen-negative and others are antigen-low1,2,4-9. Unlike the mechanisms that result in complete and permanent antigen loss6,8,9, those that lead to escape of antigen-low tumours remain unclear. Here, using mouse models of leukaemia, we show that CARs provoke reversible antigen loss through trogocytosis, an active process in which the target antigen is transferred to T cells, thereby decreasing target density on tumour cells and abating T cell activity by promoting fratricide T cell killing and T cell exhaustion. These mechanisms affect both CD28- and 4-1BB-based CARs, albeit differentially, depending on antigen density. These dynamic features can be offset by cooperative killing and combinatorial targeting to augment tumour responses to immunotherapy.


Subject(s)
Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Leukemia/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Escape/immunology , 4-1BB Ligand/immunology , Animals , CD28 Antigens/immunology , Cytotoxicity, Immunologic , Female , Immunotherapy, Adoptive , Leukemia/pathology , Male , Mice , Mice, Inbred NOD , Neoplasm Recurrence, Local/immunology , T-Lymphocytes/cytology
9.
Nature ; 572(7771): 665-669, 2019 08.
Article in English | MEDLINE | ID: mdl-31435014

ABSTRACT

Intestinal commensal bacteria can inhibit dense colonization of the gut by vancomycin-resistant Enterococcus faecium (VRE), a leading cause of hospital-acquired infections1,2. A four-strained consortium of commensal bacteria that contains Blautia producta BPSCSK can reverse antibiotic-induced susceptibility to VRE infection3. Here we show that BPSCSK reduces growth of VRE by secreting a lantibiotic that is similar to the nisin-A produced by Lactococcus lactis. Although the growth of VRE is inhibited by BPSCSK and L. lactis in vitro, only BPSCSK colonizes the colon and reduces VRE density in vivo. In comparison to nisin-A, the BPSCSK lantibiotic has reduced activity against intestinal commensal bacteria. In patients at high risk of VRE infection, high abundance of the lantibiotic gene is associated with reduced density of E. faecium. In germ-free mice transplanted with patient-derived faeces, resistance to VRE colonization correlates with abundance of the lantibiotic gene. Lantibiotic-producing commensal strains of the gastrointestinal tract reduce colonization by VRE and represent potential probiotic agents to re-establish resistance to VRE.


Subject(s)
Bacteriocins/metabolism , Bacteriocins/pharmacology , Enterococcus faecium/drug effects , Lactococcus lactis/metabolism , Probiotics , Vancomycin Resistance/drug effects , Vancomycin-Resistant Enterococci/drug effects , Animals , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacteriocins/genetics , Bacteriocins/isolation & purification , Enterococcus faecium/growth & development , Enterococcus faecium/isolation & purification , Feces/microbiology , Female , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Germ-Free Life , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/growth & development , Humans , Lactococcus lactis/chemistry , Lactococcus lactis/growth & development , Lactococcus lactis/physiology , Mice , Microbial Sensitivity Tests , Microbiota/genetics , Nisin/chemistry , Nisin/pharmacology , Symbiosis/drug effects , Vancomycin/pharmacology , Vancomycin-Resistant Enterococci/growth & development , Vancomycin-Resistant Enterococci/isolation & purification
10.
Proc Natl Acad Sci U S A ; 119(37): e2203738119, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36067288

ABSTRACT

Long noncoding RNAs (lncRNAs) play essential roles in the development and progression of many cancers. However, the contributions of lncRNAs to medulloblastoma (MB) remain poorly understood. Here, we identify Miat as an lncRNA enriched in the sonic hedgehog group of MB that is required for maintenance of a treatment-resistant stem-like phenotype in the disease. Loss of Miat results in the differentiation of tumor-initiating, stem-like MB cells and enforces the differentiation of tumorigenic stem-like MB cells into a nontumorigenic state. Miat expression in stem-like MB cells also facilitates treatment resistance by down-regulating p53 signaling and impairing radiation-induced cell death, which can be reversed by therapeutic inhibition of Miat using antisense oligonucleotides. Mechanistically, the RNA binding protein Metadherin (Mtdh), previously linked to resistance to cytotoxic therapy in cancer, binds to Miat in stem-like MB cells. Like the loss of Miat, the loss of Mtdh reduces tumorigenicity and increases sensitivity to radiation-induced death in stem-like MB cells. Moreover, Miat and Mtdh function to regulate the biogenesis of several microRNAs and facilitate tumorigenesis and treatment resistance. Taken together, these data reveal an essential role for the lncRNA Miat in sustaining a treatment-resistant pool of tumorigenic stem-like MB cells.


Subject(s)
Carcinogenesis , Cerebellar Neoplasms , Medulloblastoma , Membrane Proteins , MicroRNAs , RNA, Long Noncoding , RNA-Binding Proteins , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Humans , Medulloblastoma/genetics , Medulloblastoma/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
11.
EMBO J ; 39(8): e103334, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32134147

ABSTRACT

The production and secretion of matrix proteins upon stimulation of fibroblasts by transforming growth factor-beta (TGFß) play a critical role in wound healing. How TGFß supports the bioenergetic cost of matrix protein synthesis is not fully understood. Here, we show that TGFß promotes protein translation at least in part by increasing the mitochondrial oxidation of glucose and glutamine carbons to support the bioenergetic demand of translation. Surprisingly, we found that in addition to stimulating the entry of glucose and glutamine carbon into the TCA cycle, TGFß induced the biosynthesis of proline from glutamine in a Smad4-dependent fashion. Metabolic manipulations that increased mitochondrial redox generation promoted proline biosynthesis, while reducing mitochondrial redox potential and/or ATP synthesis impaired proline biosynthesis. Thus, proline biosynthesis acts as a redox vent, preventing the TGFß-induced increase in mitochondrial glucose and glutamine catabolism from generating damaging reactive oxygen species (ROS) when TCA cycle activity exceeds the ability of oxidative phosphorylation to convert mitochondrial redox potential into ATP. In turn, the enhanced synthesis of proline supports TGFß-induced production of matrix proteins.


Subject(s)
Fibrosis/metabolism , Glucose/metabolism , Glutamine/metabolism , Mitochondria/metabolism , Proline/metabolism , Transforming Growth Factor beta/metabolism , Animals , Citric Acid Cycle , Collagen/metabolism , Energy Metabolism , Humans , Mice , NIH 3T3 Cells , Oxidation-Reduction , Oxidative Phosphorylation , Reactive Oxygen Species/metabolism
14.
Hum Mol Genet ; 29(7): 1168-1179, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32160276

ABSTRACT

Glutaric aciduria type 1 (GA1) is an inborn error of lysine degradation characterized by a specific encephalopathy that is caused by toxic accumulation of lysine degradation intermediates. Substrate reduction through inhibition of DHTKD1, an enzyme upstream of the defective glutaryl-CoA dehydrogenase, has been investigated as a potential therapy, but revealed the existence of an alternative enzymatic source of glutaryl-CoA. Here, we show that loss of DHTKD1 in glutaryl-CoA dehydrogenase-deficient HEK-293 cells leads to a 2-fold decrease in the established GA1 clinical biomarker glutarylcarnitine and demonstrate that oxoglutarate dehydrogenase (OGDH) is responsible for this remaining glutarylcarnitine production. We furthermore show that DHTKD1 interacts with OGDH, dihydrolipoyl succinyltransferase and dihydrolipoamide dehydrogenase to form a hybrid 2-oxoglutaric and 2-oxoadipic acid dehydrogenase complex. In summary, 2-oxoadipic acid is a substrate for DHTKD1, but also for OGDH in a cell model system. The classical 2-oxoglutaric dehydrogenase complex can exist as a previously undiscovered hybrid containing DHTKD1 displaying improved kinetics towards 2-oxoadipic acid.


Subject(s)
Acyl Coenzyme A/genetics , Amino Acid Metabolism, Inborn Errors/genetics , Brain Diseases, Metabolic/genetics , Glutaryl-CoA Dehydrogenase/deficiency , Ketoglutarate Dehydrogenase Complex/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/pathology , Brain Diseases, Metabolic/metabolism , Brain Diseases, Metabolic/pathology , Cells, Cultured , Glutaryl-CoA Dehydrogenase/genetics , Glutaryl-CoA Dehydrogenase/metabolism , HEK293 Cells , Humans , Ketone Oxidoreductases/genetics , Substrate Specificity/genetics
15.
Nat Chem Biol ; 14(3): 206-214, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29443976

ABSTRACT

Despite decades of accumulated knowledge about proteins and their post-translational modifications (PTMs), numerous questions remain regarding their molecular composition and biological function. One of the most fundamental queries is the extent to which the combinations of DNA-, RNA- and PTM-level variations explode the complexity of the human proteome. Here, we outline what we know from current databases and measurement strategies including mass spectrometry-based proteomics. In doing so, we examine prevailing notions about the number of modifications displayed on human proteins and how they combine to generate the protein diversity underlying health and disease. We frame central issues regarding determination of protein-level variation and PTMs, including some paradoxes present in the field today. We use this framework to assess existing data and to ask the question, "How many distinct primary structures of proteins (proteoforms) are created from the 20,300 human genes?" We also explore prospects for improving measurements to better regularize protein-level biology and efficiently associate PTMs to function and phenotype.


Subject(s)
Genome, Human , Protein Processing, Post-Translational , Proteins/chemistry , Proteome/chemistry , Proteomics/methods , Databases, Protein , Humans , Mass Spectrometry , Phenotype , Protein Biosynthesis , Protein Isoforms/chemistry , Ubiquitin/chemistry
16.
J Biol Chem ; 292(33): 13507-13513, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28655768

ABSTRACT

Wnt proteins are a family of secreted signaling proteins that play key roles in regulating cell proliferation in both embryonic and adult tissues. Production of active Wnt depends on attachment of palmitoleate, a monounsaturated fatty acid, to a conserved serine by the acyltransferase Porcupine (PORCN). Studies of PORCN activity relied on cell-based fatty acylation and signaling assays as no direct enzyme assay had yet been developed. Here, we present the first in vitro assay that accurately recapitulates PORCN-mediated fatty acylation of a Wnt substrate. The critical feature is the use of a double disulfide-bonded Wnt peptide that mimics the two-dimensional structure surrounding the Wnt acylation site. PORCN-mediated Wnt acylation was abolished when the Wnt peptide was treated with DTT, and did not occur with a linear (non-disulfide-bonded) peptide, or when the double disulfide-bonded Wnt peptide contained Ala substituted for the Ser acylation site. We exploited this in vitro Wnt acylation assay to provide direct evidence that the small molecule LGK974, which is in clinical trials for managing Wnt-driven tumors, is a bona fide PORCN inhibitor whose IC50 for inhibition of Wnt fatty acylation in vitro closely matches that for inhibition of Wnt signaling. Side-by-side comparison of PORCN and Hedgehog acyltransferase (HHAT), two enzymes that attach 16-carbon fatty acids to secreted proteins, revealed that neither enzyme will accept the other's fatty acyl-CoA or peptide substrates. These findings illustrate the unique enzyme-substrate selectivity exhibited by members of the membrane-bound O-acyl transferase family.


Subject(s)
Acyltransferases/metabolism , Focal Dermal Hypoplasia/genetics , Membrane Proteins/metabolism , Point Mutation , Protein Processing, Post-Translational , Wnt3A Protein/metabolism , Acylation/drug effects , Acyltransferases/antagonists & inhibitors , Acyltransferases/chemistry , Acyltransferases/genetics , Amino Acid Substitution , Animals , Cystine/chemistry , Cystine/metabolism , Enzyme Inhibitors/pharmacology , Focal Dermal Hypoplasia/metabolism , HEK293 Cells , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Conformation , Protein Processing, Post-Translational/drug effects , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Substrate Specificity , Wnt Signaling Pathway/drug effects , Wnt3A Protein/chemistry
17.
Proc Natl Acad Sci U S A ; 112(11): E1220-9, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25733866

ABSTRACT

The gain-of-function mutant p53 (mtp53) transcriptome has been studied, but, to date, no detailed analysis of the mtp53-associated proteome has been described. We coupled cell fractionation with stable isotope labeling with amino acids in cell culture (SILAC) and inducible knockdown of endogenous mtp53 to determine the mtp53-driven proteome. Our fractionation data highlight the underappreciated biology that missense mtp53 proteins R273H, R280K, and L194F are tightly associated with chromatin. Using SILAC coupled to tandem MS, we identified that R273H mtp53 expression in MDA-MB-468 breast cancer cells up- and down-regulated multiple proteins and metabolic pathways. Here we provide the data set obtained from sequencing 73,154 peptide pairs that then corresponded to 3,010 proteins detected under reciprocal labeling conditions. Importantly, the high impact regulated targets included the previously identified transcriptionally regulated mevalonate pathway proteins but also identified two new levels of mtp53 protein regulation for nontranscriptional targets. Interestingly, mtp53 depletion profoundly influenced poly(ADP ribose) polymerase 1 (PARP1) localization, with increased cytoplasmic and decreased chromatin-associated protein. An enzymatic PARP shift occurred with high mtp53 expression, resulting in increased poly-ADP-ribosylated proteins in the nucleus. Mtp53 increased the level of proliferating cell nuclear antigen (PCNA) and minichromosome maintenance 4 (MCM4) proteins without changing the amount of pcna and mcm4 transcripts. Pathway enrichment analysis ranked the DNA replication pathway above the cholesterol biosynthesis pathway as a R273H mtp53 activated proteomic target. Knowledge of the proteome diversity driven by mtp53 suggests that DNA replication and repair pathways are major targets of mtp53 and highlights consideration of combination chemotherapeutic strategies targeting cholesterol biosynthesis and PARP inhibition.


Subject(s)
Breast Neoplasms/metabolism , Minichromosome Maintenance Complex Component 4/metabolism , Mutant Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Proteome/metabolism , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chromatin/drug effects , Chromatin/metabolism , Cytoplasm/drug effects , Cytoplasm/metabolism , DNA Replication/drug effects , Enzyme Inhibitors/pharmacology , Female , Humans , Isotope Labeling , Mevalonic Acid/metabolism , Protein Stability/drug effects , Protein Transport/drug effects , Proteomics , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Tumor Suppressor Protein p53/metabolism
18.
J Biol Chem ; 291(11): 6026-6035, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26801615

ABSTRACT

Autophagy is a conserved catabolic process that utilizes a defined series of membrane trafficking events to generate a de novo double-membrane vesicle termed the autophagosome, which matures by fusing to the lysosome. Subsequently, the lysosome facilitates the degradation and recycling of the cytoplasmic cargo. In yeast, the upstream signals that regulate the induction of starvation-induced autophagy are clearly defined. The nutrient-sensing kinase Tor inhibits the activation of autophagy by regulating the formation of the Atg1-Atg13-Atg17 complex, through hyperphosphorylation of Atg13. However, in mammals, the ortholog complex ULK1-ATG13-FIP200 is constitutively formed. As such, the molecular mechanism by which mTOR regulates mammalian autophagy is unknown. Here we report the identification and characterization of novel nutrient-regulated phosphorylation sites on ATG13: Ser-224 and Ser-258. mTOR directly phosphorylates ATG13 on Ser-258 while Ser-224 is modulated by the AMPK pathway. In ATG13 knock-out cells reconstituted with an unphosphorylatable mutant of ATG13, ULK1 kinase activity is more potent, and amino acid starvation induced more rapid ATG13 and ULK1 translocation. These events culminated in a more rapid starvation-induced autophagy response. Therefore, ATG13 phosphorylation plays a crucial role in autophagy regulation.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy , Starvation/metabolism , AMP-Activated Protein Kinases/metabolism , Amino Acid Sequence , Amino Acids/metabolism , Animals , Apoptosis Regulatory Proteins/chemistry , Cell Line , HEK293 Cells , Humans , Mice , Molecular Sequence Data , Phosphorylation , Sequence Alignment , TOR Serine-Threonine Kinases/metabolism
19.
J Am Chem Soc ; 139(22): 7632-7639, 2017 06 07.
Article in English | MEDLINE | ID: mdl-28448128

ABSTRACT

The Ras proteins are essential GTPases involved in the regulation of cell proliferation and survival. Mutated oncogenic forms of Ras alter effector binding and innate GTPase activity, leading to deregulation of downstream signal transduction. Mutated forms of Ras are involved in approximately 30% of human cancers. Despite decades of effort to develop direct Ras inhibitors, Ras has long been considered "undruggable" due to its high affinity for GTP and its lack of hydrophobic binding pockets. Herein, we report a total chemical synthesis of all-l- and all-d-amino acid biotinylated variants of oncogenic mutant KRas(G12V). The protein is synthesized using Fmoc-based solid-phase peptide synthesis and assembled using combined native chemical ligation and isonitrile-mediated activation strategies. We demonstrate that both KRas(G12V) enantiomers can successfully fold and bind nucleotide substrates and binding partners with observable enantiodiscrimination. By demonstrating the functional competency of a mirror-image form of KRas bound to its corresponding enantiomeric nucleotide triphosphate, this study sets the stage for further biochemical studies with this material. In particular, this protein will enable mirror-image yeast surface display experiments to identify all-d peptide ligands for oncogenic KRas, providing a useful tool in the search for new therapeutics against this challenging disease target.


Subject(s)
Proto-Oncogene Proteins p21(ras)/chemical synthesis , Amino Acid Sequence , Genetic Variation , Humans , Protein Folding , Proto-Oncogene Proteins p21(ras)/genetics
20.
J Am Chem Soc ; 137(40): 13167-75, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26401918

ABSTRACT

Human granulocyte colony-stimulating factor (G-CSF) is an endogenous glycoprotein involved in hematopoiesis. Natively glycosylated and nonglycosylated recombinant forms, lenograstim and filgrastim, respectively, are used clinically to manage neutropenia in patients undergoing chemotherapeutic treatment. Despite their comparable therapeutic potential, the purpose of O-linked glycosylation at Thr133 remains a subject of controversy. In light of this, we have developed a synthetic platform to prepare G-CSF aglycone with the goal of enabling access to native and designed glycoforms with site-selectivity and glycan homogeneity. To address the synthesis of a relatively large, aggregation-prone sequence, we advanced an isonitrile-mediated ligation method. The chemoselective activation and coupling of C-terminal peptidyl Gly thioacids with the N-terminus of an unprotected peptide provide ligated peptides directly in a manner complementary to that with conventional native chemical ligation-desulfurization strategies. Herein, we describe the details and application of this method as it enabled the convergent total synthesis of G-CSF aglycone.


Subject(s)
Granulocyte Colony-Stimulating Factor/chemistry , Peptides/chemistry , Amino Acid Sequence , Electrophoresis, Polyacrylamide Gel , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL