Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell ; 186(24): 5328-5346.e26, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37883971

ABSTRACT

Lysosomes serve dual antagonistic functions in cells by mediating anabolic growth signaling and the catabolic turnover of macromolecules. How these janus-faced activities are regulated in response to cellular nutrient status is poorly understood. We show here that lysosome morphology and function are reversibly controlled by a nutrient-regulated signaling lipid switch that triggers the conversion between peripheral motile mTOR complex 1 (mTORC1) signaling-active and static mTORC1-inactive degradative lysosomes clustered at the cell center. Starvation-triggered relocalization of phosphatidylinositol 4-phosphate (PI(4)P)-metabolizing enzymes reshapes the lysosomal surface proteome to facilitate lysosomal proteolysis and to repress mTORC1 signaling. Concomitantly, lysosomal phosphatidylinositol 3-phosphate (PI(3)P), which marks motile signaling-active lysosomes in the cell periphery, is erased. Interference with this PI(3)P/PI(4)P lipid switch module impairs the adaptive response of cells to altering nutrient supply. Our data unravel a key function for lysosomal phosphoinositide metabolism in rewiring organellar membrane dynamics in response to cellular nutrient status.


Subject(s)
Lysosomes , Signal Transduction , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Nutrients , Cell Physiological Phenomena
2.
Elife ; 122023 10 16.
Article in English | MEDLINE | ID: mdl-37843983

ABSTRACT

Inositol hexakisphosphate kinases (IP6Ks) are emerging as relevant pharmacological targets because a multitude of disease-related phenotypes has been associated with their function. While the development of potent IP6K inhibitors is gaining momentum, a pharmacological tool to distinguish the mammalian isozymes is still lacking. Here, we implemented an analog-sensitive approach for IP6Ks and performed a high-throughput screen to identify suitable lead compounds. The most promising hit, FMP-201300, exhibited high potency and selectivity toward the unique valine gatekeeper mutants of IP6K1 and IP6K2, compared to the respective wild-type (WT) kinases. Biochemical validation experiments revealed an allosteric mechanism of action that was corroborated by hydrogen deuterium exchange mass spectrometry measurements. The latter analysis suggested that displacement of the αC helix, caused by the gatekeeper mutation, facilitates the binding of FMP-201300 to an allosteric pocket adjacent to the ATP-binding site. FMP-201300 therefore serves as a valuable springboard for the further development of compounds that can selectively target the three mammalian IP6Ks; either as analog-sensitive kinase inhibitors or as an allosteric lead compound for the WT kinases.


Subject(s)
Phosphotransferases (Phosphate Group Acceptor) , Phytic Acid , Animals , Phosphotransferases (Phosphate Group Acceptor)/genetics , Phosphotransferases (Phosphate Group Acceptor)/metabolism , Inositol Phosphates/metabolism , Mammals/metabolism
3.
Chembiochem ; 24(24): e202300555, 2023 12 14.
Article in English | MEDLINE | ID: mdl-37769151

ABSTRACT

Uridine diphosphate N-acetylglucosamine 2-epimerase (GNE) is a key enzyme in the sialic acid biosynthesis pathway. Sialic acids are primarily terminal carbohydrates on glycans and play fundamental roles in health and disease. In search of effective GNE inhibitors not based on a carbohydrate scaffold, we performed a high-throughput screening campaign of 68,640 drug-like small molecules against recombinant GNE using a UDP detection assay. We validated nine of the primary actives with an orthogonal real-time NMR assay and verified their IC50 values in the low micromolar to nanomolar range manually. Stability and solubility studies revealed three compounds for further evaluation. Thermal shift assays, analytical size exclusion, and interferometric scattering microscopy demonstrated that the GNE inhibitors acted on the oligomeric state of the protein. Finally, hydrogen-deuterium exchange mass spectrometry (HDX-MS) revealed which sections of GNE were shifted upon the addition of the inhibitors. In summary, we have identified three small molecules as GNE inhibitors with high potency in vitro, which serve as promising candidates to modulate sialic acid biosynthesis in more complex systems.


Subject(s)
Carbohydrate Epimerases , N-Acetylneuraminic Acid , Humans , Carbohydrate Epimerases/chemistry , Carbohydrate Epimerases/metabolism , Sialic Acids/chemistry , Carbohydrates , Polysaccharides
4.
Structure ; 28(2): 145-156.e5, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31831213

ABSTRACT

The class I phosphoinositide 3-kinases (PI3Ks) are key signaling enzymes composed of a heterodimer of a p110 catalytic subunit and a p85 regulatory subunit, with PI3K mutations being causative of multiple human diseases including cancer, primary immunodeficiencies, and developmental disorders. Mutations in the p85α regulatory subunit encoded by PIK3R1 can both activate PI3K through oncogenic truncations in the iSH2 domain, or inhibit PI3K through developmental disorder mutations in the cSH2 domain. Using a combined biochemical and hydrogen deuterium exchange mass spectrometry approach we have defined the molecular basis for how these mutations alter the activity of p110α/p110δ catalytic subunits. We find that the oncogenic Q572∗ truncation of PIK3R1 disrupts all p85-inhibitory inputs, with p110α being hyper-activated compared with p110δ. In addition, we find that the R649W mutation in the cSH2 of PIK3R1 decreases sensitivity to activation by receptor tyrosine kinases. This work reveals unique insight into isoform-specific regulation of p110s by p85α.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , Class Ia Phosphatidylinositol 3-Kinase/genetics , Mutation , Catalytic Domain , Class I Phosphatidylinositol 3-Kinases/chemistry , Class Ia Phosphatidylinositol 3-Kinase/chemistry , Enzyme Activation , Humans , Hydrogen Deuterium Exchange-Mass Spectrometry , Models, Molecular , Protein Conformation , Protein Domains
5.
J Mol Biol ; 430(18 Pt B): 3129-3142, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30031006

ABSTRACT

Phosphatidylinositol 4-kinase IIIα (PI4KIIIα) is the lipid kinase primarily responsible for generating the lipid phosphatidylinositol 4-phosphate (PI4P) at the plasma membrane, which acts as the substrate for generation of the signaling lipids PIP2 and PIP3. PI4KIIIα forms a large heterotrimeric complex with two regulatory partners, TTC7 and FAM126. We describe using an integrated electron microscopy and hydrogen-deuterium exchange mass spectrometry (HDX-MS) approach to probe the architecture and dynamics of the complex of PI4KIIIα/TTC7/FAM126. HDX-MS reveals that the majority of the PI4KIIIα sequence was protected from exchange in short deuterium pulse experiments, suggesting presence of secondary structure, even in putative unstructured regions. Negative stain electron microscopy reveals the shape and architecture of the full-length complex, revealing an overall dimer of PI4KIIIα/TTC7/FAM126 trimers. HDX-MS reveals conformational changes in the TTC7/FAM126 complex upon binding PI4KIIIα, including both at the direct TTC7-PI4KIIIα interface and at the putative membrane binding surface. Finally, HDX-MS experiments of PI4KIIIα bound to the highly potent and selective inhibitor GSK-A1 compared to that bound to the non-specific inhibitor PIK93 revealed substantial conformational changes throughout an extended region of the kinase domain. Many of these changes were distant from the putative inhibitor binding site, showing a large degree of allosteric conformational changes that occur upon inhibitor binding. Overall, our results reveal novel insight into the regulation of PI4KIIIα by its regulatory proteins TTC7/FAM126, as well as additional dynamic information on how selective inhibition of PI4KIIIα is achieved.


Subject(s)
1-Phosphatidylinositol 4-Kinase/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes/metabolism , Proteins/metabolism , 1-Phosphatidylinositol 4-Kinase/chemistry , Allosteric Regulation , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , Protein Binding , Protein Conformation , Protein Multimerization , Proteins/chemistry , Recombinant Proteins
6.
Front Immunol ; 9: 575, 2018.
Article in English | MEDLINE | ID: mdl-29616047

ABSTRACT

The signaling lipid phosphatidylinositol 3,4,5, trisphosphate (PIP3) is an essential mediator of many vital cellular processes, including growth, survival, and metabolism. PIP3 is generated through the action of the class I phosphoinositide 3-kinases (PI3K), and their activity is tightly controlled through interactions with regulatory proteins and activating stimuli. The class IA PI3Ks are composed of three distinct p110 catalytic subunits (p110α, p110ß, and p110δ), and they play different roles in specific tissues due to disparities in both expression and engagement downstream of cell-surface receptors. Disruption of PI3K regulation is a frequent driver of numerous human diseases. Activating mutations in the PIK3CA gene encoding the p110α catalytic subunit of class IA PI3K are frequently mutated in several cancer types, and mutations in the PIK3CD gene encoding the p110δ catalytic subunit have been identified in primary immunodeficiency patients. All class IA p110 subunits interact with p85 regulatory subunits, and mutations/deletions in different p85 regulatory subunits have been identified in both cancer and primary immunodeficiencies. In this review, we will summarize our current understanding for the molecular basis of how class IA PI3K catalytic activity is regulated by p85 regulatory subunits, and how activating mutations in the PI3K catalytic subunits PIK3CA and PIK3CD (p110α, p110δ) and regulatory subunits PIK3R1 (p85α) mediate PI3K activation and human disease.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/genetics , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Disease Susceptibility , Immunologic Deficiency Syndromes/etiology , Immunologic Deficiency Syndromes/metabolism , Mutation , Neoplasms/etiology , Neoplasms/metabolism , Oncogenes , Animals , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Developmental Disabilities/etiology , Developmental Disabilities/metabolism , Gene Expression Regulation , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
7.
Science ; 358(6366): 1056-1059, 2017 11 24.
Article in English | MEDLINE | ID: mdl-29074584

ABSTRACT

Newly transcribed eukaryotic precursor messenger RNAs (pre-mRNAs) are processed at their 3' ends by the ~1-megadalton multiprotein cleavage and polyadenylation factor (CPF). CPF cleaves pre-mRNAs, adds a polyadenylate tail, and triggers transcription termination, but it is unclear how its various enzymes are coordinated and assembled. Here, we show that the nuclease, polymerase, and phosphatase activities of yeast CPF are organized into three modules. Using electron cryomicroscopy, we determined a 3.5-angstrom-resolution structure of the ~200-kilodalton polymerase module. This revealed four ß propellers, in an assembly markedly similar to those of other protein complexes that bind nucleic acid. Combined with in vitro reconstitution experiments, our data show that the polymerase module brings together factors required for specific and efficient polyadenylation, to help coordinate mRNA 3'-end processing.


Subject(s)
RNA 3' End Processing , RNA Polymerase II/chemistry , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , mRNA Cleavage and Polyadenylation Factors/chemistry , Cryoelectron Microscopy , Polynucleotide Adenylyltransferase/metabolism , Protein Conformation , RNA Polymerase II/ultrastructure , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/ultrastructure , mRNA Cleavage and Polyadenylation Factors/ultrastructure
9.
Proc Natl Acad Sci U S A ; 114(8): 1982-1987, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28167755

ABSTRACT

Activated PI3K Delta Syndrome (APDS) is a primary immunodeficiency disease caused by activating mutations in either the leukocyte-restricted p110δ catalytic (PIK3CD) subunit or the ubiquitously expressed p85α regulatory (PIK3R1) subunit of class IA phosphoinositide 3-kinases (PI3Ks). There are two classes of APDS: APDS1 that arises from p110δ mutations that are analogous to oncogenic mutations found in the broadly expressed p110α subunit and APDS2 that occurs from a splice mutation resulting in p85α with a central deletion (Δ434-475). As p85 regulatory subunits associate with and inhibit all class IA catalytic subunits, APDS2 mutations are expected to similarly activate p110α, ß, and δ, yet APDS2 largely phenocopies APDS1 without dramatic effects outside the immune system. We have examined the molecular mechanism of activation of both classes of APDS mutations using a combination of biochemical assays and hydrogen-deuterium exchange mass spectrometry. Intriguingly, we find that an APDS2 mutation in p85α leads to substantial basal activation of p110δ (>300-fold) and disrupts inhibitory interactions from the nSH2, iSH2, and cSH2 domains of p85, whereas p110α is only minimally basally activated (∼2-fold) when associated with mutated p85α. APDS1 mutations in p110δ (N334K, E525K, E1021K) mimic the activation mechanisms previously discovered for oncogenic mutations in p110α. All APDS mutations were potently inhibited by the Food and Drug Administration-approved p110δ inhibitor idelalisib. Our results define the molecular basis of how PIK3CD and PIK3R1 mutations result in APDS and reveal a potential path to treatment for all APDS patients.


Subject(s)
Catalytic Domain/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Enzyme Inhibitors/pharmacology , Immunologic Deficiency Syndromes/genetics , Phosphatidylinositol 3-Kinases/genetics , Purines/pharmacology , Quinazolinones/pharmacology , Cell Membrane/metabolism , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/metabolism , Class Ia Phosphatidylinositol 3-Kinase , Enzyme Assays , Enzyme Inhibitors/therapeutic use , Gain of Function Mutation , Humans , Immunologic Deficiency Syndromes/drug therapy , Mass Spectrometry/methods , Models, Molecular , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Primary Immunodeficiency Diseases , Protein Conformation , Purines/therapeutic use , Quinazolinones/therapeutic use , Randomized Controlled Trials as Topic , Sequence Deletion
10.
Nature ; 538(7625): 344-349, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27602946

ABSTRACT

Antimalarial drugs have thus far been chiefly derived from two sources-natural products and synthetic drug-like compounds. Here we investigate whether antimalarial agents with novel mechanisms of action could be discovered using a diverse collection of synthetic compounds that have three-dimensional features reminiscent of natural products and are underrepresented in typical screening collections. We report the identification of such compounds with both previously reported and undescribed mechanisms of action, including a series of bicyclic azetidines that inhibit a new antimalarial target, phenylalanyl-tRNA synthetase. These molecules are curative in mice at a single, low dose and show activity against all parasite life stages in multiple in vivo efficacy models. Our findings identify bicyclic azetidines with the potential to both cure and prevent transmission of the disease as well as protect at-risk populations with a single oral dose, highlighting the strength of diversity-oriented synthesis in revealing promising therapeutic targets.


Subject(s)
Antimalarials/chemical synthesis , Antimalarials/pharmacology , Azetidines/therapeutic use , Drug Discovery , Life Cycle Stages/drug effects , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development , Animals , Antimalarials/administration & dosage , Antimalarials/therapeutic use , Azabicyclo Compounds/administration & dosage , Azabicyclo Compounds/chemical synthesis , Azabicyclo Compounds/pharmacology , Azabicyclo Compounds/therapeutic use , Azetidines/administration & dosage , Azetidines/adverse effects , Azetidines/pharmacology , Cytosol/enzymology , Disease Models, Animal , Female , Liver/drug effects , Liver/parasitology , Macaca mulatta/parasitology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Male , Mice , Phenylalanine-tRNA Ligase/antagonists & inhibitors , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/chemical synthesis , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Plasmodium falciparum/cytology , Plasmodium falciparum/enzymology , Safety
11.
J Med Chem ; 59(5): 1830-9, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26885694

ABSTRACT

Type III phosphatidylinositol 4-kinase (PI4KIIIß) is an essential enzyme in mediating membrane trafficking and is implicated in a variety of pathogenic processes. It is a key host factor mediating replication of RNA viruses. The design of potent and specific inhibitors of this enzyme will be essential to define its cellular roles and may lead to novel antiviral therapeutics. We previously reported the PI4K inhibitor PIK93, and this compound has defined key functions of PI4KIIIß. However, this compound showed high cross reactivity with class I and III PI3Ks. Using structure-based drug design, we have designed novel potent and selective (>1000-fold over class I and class III PI3Ks) PI4KIIIß inhibitors. These compounds showed antiviral activity against hepatitis C virus. The co-crystal structure of PI4KIIIß bound to one of the most potent compounds reveals the molecular basis of specificity. This work will be vital in the design of novel PI4KIIIß inhibitors, which may play significant roles as antiviral therapeutics.


Subject(s)
Antiviral Agents/pharmacology , Drug Design , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Hepacivirus/drug effects , Hepacivirus/physiology , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinase Inhibitors/chemical synthesis , Structure-Activity Relationship , Virus Replication/drug effects
12.
Biochem Soc Trans ; 44(1): 260-6, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26862213

ABSTRACT

Many important cellular functions are regulated by the selective recruitment of proteins to intracellular membranes mediated by specific interactions with lipid phosphoinositides. The enzymes that generate lipid phosphoinositides therefore must be properly positioned and regulated at their correct cellular locations. Phosphatidylinositol 4 kinases (PI4Ks) are key lipid signalling enzymes, and they generate the lipid species phosphatidylinositol 4-phosphate (PI4P), which plays important roles in regulating physiological processes including membrane trafficking, cytokinesis and organelle identity. PI4P also acts as the substrate for the generation of the signalling phosphoinositides phosphatidylinositol 4,5-bisphosphate (PIP2) and phosphatidylinositol 3,4,5-trisphosphate (PIP3). PI4Ks also play critical roles in a number of pathological processes including mediating replication of a number of pathogenic RNA viruses, and in the development of the parasite responsible for malaria. Key to the regulation of PI4Ks is their regulation by a variety of both host and viral protein-binding partners. We review herein our current understanding of the structure, regulatory interactions and role in disease of the type III PI4Ks.


Subject(s)
1-Phosphatidylinositol 4-Kinase/chemistry , 1-Phosphatidylinositol 4-Kinase/metabolism , Disease , Signal Transduction , Animals , Humans , Models, Molecular , Protein Binding
13.
Mol Cell ; 54(5): 858-69, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24905007

ABSTRACT

Fanconi anaemia (FA) is a cancer predisposition syndrome characterized by cellular sensitivity to DNA interstrand crosslinkers. The molecular defect in FA is an impaired DNA repair pathway. The critical event in activating this pathway is monoubiquitination of FANCD2. In vivo, a multisubunit FA core complex catalyzes this step, but its mechanism is unclear. Here, we report purification of a native avian FA core complex and biochemical reconstitution of FANCD2 monoubiquitination. This demonstrates that the catalytic FANCL E3 ligase subunit must be embedded within the complex for maximal activity and site specificity. We genetically and biochemically define a minimal subcomplex comprising just three proteins (FANCB, FANCL, and FAAP100) that functions as the monoubiquitination module. Residual FANCD2 monoubiquitination activity is retained in cells defective for other FA core complex subunits. This work describes the in vitro reconstitution and characterization of this multisubunit monoubiquitin E3 ligase, providing key insight into the conserved FA DNA repair pathway.


Subject(s)
Avian Proteins/metabolism , Chickens/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Ubiquitination , Animals , Avian Proteins/chemistry , Avian Proteins/genetics , Cell Line , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group D2 Protein/chemistry , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group L Protein/chemistry , Fanconi Anemia Complementation Group L Protein/metabolism , Fanconi Anemia Complementation Group Proteins/chemistry , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Humans , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...