Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Nat Cell Biol ; 24(9): 1422-1432, 2022 09.
Article in English | MEDLINE | ID: mdl-36050469

ABSTRACT

Tumour dependency on specific metabolic signals has been demonstrated and often guided numerous therapeutic approaches. We identify melanoma addiction to the mitochondrial protein glutaryl-CoA dehydrogenase (GCDH), which functions in lysine metabolism and controls protein glutarylation. GCDH knockdown induced cell death programmes in melanoma cells, an activity blocked by inhibition of the upstream lysine catabolism enzyme DHTKD1. The transcription factor NRF2 mediates GCDH-dependent melanoma cell death programmes. Mechanistically, GCDH knockdown induces NRF2 glutarylation, increasing its stability and DNA binding activity, with a concomitant transcriptional upregulation of ATF4, ATF3, DDIT3 and CHAC1, resulting in cell death. In vivo, inducible inactivation of GCDH effectively inhibited melanoma tumour growth. Correspondingly, reduced GCDH expression correlated with improved survival of patients with melanoma. These findings identify melanoma cell addiction to GCDH, limiting apoptotic signalling by controlling NRF2 glutarylation. Inhibiting the GCDH pathway could thus represent a therapeutic approach to treat melanoma.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Brain Diseases, Metabolic , Melanoma , NF-E2-Related Factor 2/metabolism , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Brain Diseases, Metabolic/genetics , Brain Diseases, Metabolic/metabolism , Brain Diseases, Metabolic/pathology , DNA , Glutaryl-CoA Dehydrogenase/genetics , Glutaryl-CoA Dehydrogenase/metabolism , Humans , Ketoglutarate Dehydrogenase Complex , Lysine , Melanoma/genetics , Mitochondrial Proteins , NF-E2-Related Factor 2/genetics
2.
Bioorg Med Chem Lett ; 41: 128007, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33798699

ABSTRACT

NAD+ is a crucial cellular factor that plays multifaceted roles in wide ranging biological processes. Low levels of NAD+ have been linked to numerous diseases including metabolic disorders, cardiovascular disease, neurodegeneration, and muscle wasting disorders. A novel strategy to boost NAD+ is to activate nicotinamide phosphoribosyltransferase (NAMPT), the putative rate-limiting step in the NAD+ salvage pathway. We previously showed that NAMPT activators increase NAD+ levels in vitro and in vivo. Herein we describe the optimization of our NAMPT activator prototype (SBI-0797812) leading to the identification of 1-(4-((4-chlorophenyl)sulfonyl)phenyl)-3-(oxazol-5-ylmethyl)urea (34) that showed far more potent NAMPT activation and improved oral bioavailability.


Subject(s)
Cytokines/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Urea/pharmacology , Dose-Response Relationship, Drug , Humans , Molecular Structure , Structure-Activity Relationship , Urea/analogs & derivatives , Urea/chemistry
3.
Cancer Res ; 81(6): 1472-1485, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33414172

ABSTRACT

Invadopodia are actin-based proteolytic membrane protrusions required for invasive behavior and tumor growth. In this study, we used our high-content screening assay to identify kinases whose activity affects invadopodia formation. Among the top hits selected for further analysis was TAO3, an STE20-like kinase of the GCK subfamily. TAO3 was overexpressed in many human cancers and regulated invadopodia formation in melanoma, breast, and bladder cancers. Furthermore, TAO3 catalytic activity facilitated melanoma growth in three-dimensional matrices and in vivo. A novel, potent catalytic inhibitor of TAO3 was developed that inhibited invadopodia formation and function as well as tumor cell extravasation and growth. Treatment with this inhibitor demonstrated that TAO3 activity is required for endosomal trafficking of TKS5α, an obligate invadopodia scaffold protein. A phosphoproteomics screen for TAO3 substrates revealed the dynein subunit protein LIC2 as a relevant substrate. Knockdown of LIC2 or expression of a phosphomimetic form promoted invadopodia formation. Thus, TAO3 is a new therapeutic target with a distinct mechanism of action. SIGNIFICANCE: An unbiased screening approach identifies TAO3 as a regulator of invadopodia formation and function, supporting clinical development of this class of target.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Endosomes/metabolism , Neoplasm Invasiveness/pathology , Podosomes/drug effects , Protein Serine-Threonine Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cytoplasmic Dyneins/genetics , Cytoplasmic Dyneins/metabolism , Datasets as Topic , Extracellular Matrix , Female , Gene Expression Profiling , Gene Knockdown Techniques , High-Throughput Screening Assays , Humans , Male , Melanoma/drug therapy , Melanoma/pathology , Mice , Neoplasm Invasiveness/prevention & control , Podosomes/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Time-Lapse Imaging , Xenograft Model Antitumor Assays
4.
Nat Commun ; 10(1): 3241, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31324777

ABSTRACT

Pharmacological strategies that boost intracellular NAD+ are highly coveted for their therapeutic potential. One approach is activation of nicotinamide phosphoribosyltransferase (NAMPT) to increase production of nicotinamide mononucleotide (NMN), the predominant NAD+ precursor in mammalian cells. A high-throughput screen for NAMPT activators and hit-to-lead campaign yielded SBI-797812, a compound that is structurally similar to active-site directed NAMPT inhibitors and blocks binding of these inhibitors to NAMPT. SBI-797812 shifts the NAMPT reaction equilibrium towards NMN formation, increases NAMPT affinity for ATP, stabilizes phosphorylated NAMPT at His247, promotes consumption of the pyrophosphate by-product, and blunts feedback inhibition by NAD+. These effects of SBI-797812 turn NAMPT into a "super catalyst" that more efficiently generates NMN. Treatment of cultured cells with SBI-797812 increases intracellular NMN and NAD+. Dosing of mice with SBI-797812 elevates liver NAD+. Small molecule NAMPT activators such as SBI-797812 are a pioneering approach to raise intracellular NAD+ and realize its associated salutary effects.


Subject(s)
Enzyme Activators/pharmacology , NAD/metabolism , Nicotinamide Mononucleotide/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Small Molecule Libraries/pharmacology , A549 Cells , Animals , Biocatalysis/drug effects , Enzyme Activators/administration & dosage , Enzyme Activators/chemistry , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Liver/drug effects , Liver/metabolism , Mice , Molecular Structure , Phosphorylation/drug effects , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/chemistry
5.
ACS Chem Biol ; 14(5): 949-958, 2019 05 17.
Article in English | MEDLINE | ID: mdl-30969758

ABSTRACT

Conventional treatments to combat the tuberculosis (TB) epidemic are falling short, thus encouraging the search for novel antitubercular drugs acting on unexplored molecular targets. Several whole-cell phenotypic screenings have delivered bioactive compounds with potent antitubercular activity. However, their cellular target and mechanism of action remain largely unknown. Further evaluation of these compounds may include their screening in search for known antitubercular drug targets hits. Here, a collection of nearly 1400 mycobactericidal compounds was screened against Mycobacterium tuberculosis NaMN adenylyltransferase ( MtNadD), a key enzyme in the biogenesis of NAD cofactor that was recently validated as a new drug target for dormant and active tuberculosis. We found three chemotypes that efficiently inhibit MtNadD in the low micromolar range in vitro. SAR and cheminformatics studies of commercially available analogues point to a series of benzimidazolium derivatives, here named N2, with bactericidal activity on different mycobacteria, including M. abscessus, multidrug-resistant M. tuberculosis, and dormant M. smegmatis. The on-target activity was supported by the increased resistance of an M. smegmatis strain overexpressing the target and by a rapid decline in NAD(H) levels. A cocrystal structure of MtNadD with N2-8 inhibitor reveals that the binding of the inhibitor induced the formation of a new quaternary structure, a dimer-of-dimers where two copies of the inhibitor occupy symmetrical positions in the dimer interface, thus paving the way for the development of a new generation of selective MtNadD bioactive inhibitors. All these results strongly suggest that pharmacological inhibition of MtNadD is an effective strategy to combat dormant and resistant Mtb strains.


Subject(s)
Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/drug effects , NAD/antagonists & inhibitors , Nicotinamide-Nucleotide Adenylyltransferase/antagonists & inhibitors , Drug Resistance, Bacterial , Microbial Sensitivity Tests , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/metabolism , NAD/biosynthesis , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Structure-Activity Relationship
6.
Cancer Lett ; 449: 145-162, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30771432

ABSTRACT

Inhibition of ubiquitin ligases with small molecule remains a very challenging task, given the lack of catalytic activity of the target and the requirement of disruption of its interactions with other proteins. Siah1/2, which are E3 ubiquitin ligases, are implicated in melanoma and prostate cancer and represent high-value drug targets. We utilized three independent screening approaches in our efforts to identify small-molecule Siah1/2 inhibitors: Affinity Selection-Mass Spectrometry, a protein thermal shift-based assay and an in silico based screen. Inhibitors were assessed for their effect on viability of melanoma and prostate cancer cultures, colony formation, prolyl-hydroxylase-HIF1α signaling, expression of selected Siah2-related transcripts, and Siah2 ubiquitin ligase activity. Several analogs were further characterized, demonstrating improved efficacy. Combination of the top hits identified in the different assays demonstrated an additive effect, pointing to complementing mechanisms that underlie each of these Siah1/2 inhibitors.


Subject(s)
Melanoma/drug therapy , Nuclear Proteins/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Small Molecule Libraries/administration & dosage , Ubiquitin-Protein Ligases/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Computer Simulation , Down-Regulation , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mass Spectrometry , Melanoma/genetics , Mice , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Small Molecule Libraries/isolation & purification , Small Molecule Libraries/pharmacology , Ubiquitin-Protein Ligases/genetics , Xenograft Model Antitumor Assays
7.
Bioorg Med Chem Lett ; 28(1): 31-34, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29174347

ABSTRACT

Tissue-nonspecific alkaline phosphatase (TNAP) is an ectoenzyme crucial for bone matrix mineralization via its ability to hydrolyze extracellular inorganic pyrophosphate (ePPi), a potent mineralization inhibitor, to phosphate (Pi). By the controlled hydrolysis of ePPi, TNAP maintains the correct ratio of Pi to ePPi and therefore enables normal skeletal and dental calcification. In other areas of the body low ePPi levels lead to the development of pathological soft-tissue calcification, which can progress to a number of disorders. TNAP inhibitors have been shown to prevent these processes via an increase of ePPi. Herein we describe the use of a whole blood assay to optimize a previously described series of TNAP inhibitors resulting in 5-((5-chloro-2-methoxyphenyl)sulfonamido)nicotinamide (SBI-425), a potent, selective and oral bioavailable compound that robustly inhibits TNAP in vivo.


Subject(s)
Alkaline Phosphatase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Niacinamide/analogs & derivatives , Niacinamide/chemistry , Sulfonamides/chemistry , Administration, Oral , Alkaline Phosphatase/metabolism , Animals , Drug Evaluation, Preclinical , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Half-Life , Inhibitory Concentration 50 , Mice , Niacinamide/metabolism , Niacinamide/pharmacokinetics , Structure-Activity Relationship , Sulfonamides/metabolism , Sulfonamides/pharmacokinetics
8.
Sci Transl Med ; 9(393)2017 06 07.
Article in English | MEDLINE | ID: mdl-28592560

ABSTRACT

Biallelic mutations in ABCC6 cause pseudoxanthoma elasticum (PXE), a disease characterized by calcification in the skin, eyes, and blood vessels. The function of ATP-binding cassette C6 (ABCC6) and the pathogenesis of PXE remain unclear. We used mouse models and patient fibroblasts to demonstrate genetic interaction and shared biochemical and cellular mechanisms underlying ectopic calcification in PXE and related disorders caused by defined perturbations in extracellular adenosine 5'-triphosphate catabolism. Under osteogenic culture conditions, ABCC6 mutant cells calcified, suggesting a provoked cell-autonomous defect. Using a conditional Abcc6 knockout mouse model, we excluded the prevailing pathogenic hypothesis that singularly invokes failure of hepatic secretion of an endocrine inhibitor of calcification. Instead, deficiency of Abcc6 in both local and distant cells was necessary to achieve the early onset and penetrant ectopic calcification observed upon constitutive gene targeting. ABCC6 mutant cells additionally had increased expression and activity of tissue-nonspecific alkaline phosphatase (TNAP), an enzyme that degrades pyrophosphate, a major inhibitor of calcification. A selective and orally bioavailable TNAP inhibitor prevented calcification in ABCC6 mutant cells in vitro and attenuated both the development and progression of calcification in Abcc6-/- mice in vivo, without the deleterious effects on bone associated with other proposed treatment strategies.


Subject(s)
Alkaline Phosphatase/antagonists & inhibitors , Calcinosis/complications , Calcinosis/enzymology , Pseudoxanthoma Elasticum/complications , Pseudoxanthoma Elasticum/enzymology , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate/metabolism , Alkaline Phosphatase/metabolism , Animals , Crosses, Genetic , Disease Models, Animal , Epistasis, Genetic , Extracellular Space/metabolism , Female , Fibroblasts/metabolism , Gene Deletion , Humans , Liver/metabolism , Male , Mice , Mice, Mutant Strains , Models, Biological , Multidrug Resistance-Associated Proteins/genetics , Mutation/genetics , Osteogenesis , Phenotype , Phosphoric Diester Hydrolases/metabolism , Pyrophosphatases/metabolism
9.
Methods Mol Biol ; 1439: 131-42, 2016.
Article in English | MEDLINE | ID: mdl-27316992

ABSTRACT

Fluorescence-based detection techniques are popular in high throughput screening due to sensitivity and cost-effectiveness. Four commonly used techniques exist, each with distinct characteristics. Fluorescence intensity assays are the simplest to run, but suffer the most from signal interference. Fluorescence polarization assays show less interference from the compounds or the instrument, but require a design that results in change of fluorophore-containing moiety size and usually have narrow assay signal window. Fluorescence resonance energy transfer (FRET) is commonly used for detecting protein-protein interactions and is constrained not by the sizes of binding partners, but rather by the distance between fluorophores. Time-resolved fluorescence resonance energy transfer (TR-FRET), an advanced modification of FRET approach utilizes special fluorophores with long-lived fluorescence and earns its place near the top of fluorescent techniques list by its performance and robustness, characterized by larger assay window and minimized compound spectral interference. TR-FRET technology can be applied in biochemical or cell-based in vitro assays with ease. It is commonly used to detect modulation of protein-protein interactions and in detection of products of biochemical reactions and cellular activities.


Subject(s)
Drug Evaluation, Preclinical/methods , Enzyme Assays/methods , Fluorescence Resonance Energy Transfer/methods , High-Throughput Screening Assays/methods , Protein Interaction Maps , Animals , Cullin Proteins/metabolism , Humans , NEDD8 Protein/metabolism , Protein Interaction Maps/drug effects , SUMO-1 Protein/metabolism
10.
J Mol Biol ; 428(11): 2430-2445, 2016 06 05.
Article in English | MEDLINE | ID: mdl-27091468

ABSTRACT

Multisite phosphorylation is required for the biological function of serine-arginine (SR) proteins, a family of essential regulators of mRNA splicing. These modifications are catalyzed by serine-arginine protein kinases (SRPKs) that phosphorylate numerous serines in arginine-serine-rich (RS) domains of SR proteins using a directional, C-to-N-terminal mechanism. The present studies explore how SRPKs govern this highly biased phosphorylation reaction and investigate biological roles of the observed directional phosphorylation mechanism. Using NMR spectroscopy with two separately expressed domains of SRSF1, we showed that several residues in the RNA-binding motif 2 interact with the N-terminal region of the RS domain (RS1). These contacts provide a structural framework that balances the activities of SRPK1 and the protein phosphatase PP1, thereby regulating the phosphoryl content of the RS domain. Disruption of the implicated intramolecular RNA-binding motif 2-RS domain interaction impairs both the directional phosphorylation mechanism and the nuclear translocation of SRSF1 demonstrating that the intrinsic phosphorylation bias is obligatory for SR protein biological function.


Subject(s)
Active Transport, Cell Nucleus/physiology , Phosphorylation/physiology , RNA Recognition Motif/physiology , RNA/metabolism , Serine-Arginine Splicing Factors/metabolism , Amino Acid Sequence , Arginine/metabolism , Humans , Nuclear Proteins/metabolism , Protein Binding/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , RNA Splicing/genetics , RNA-Binding Proteins/metabolism , Serine/metabolism
11.
Cell Chem Biol ; 23(4): 517-28, 2016 04 21.
Article in English | MEDLINE | ID: mdl-27105284

ABSTRACT

The AAA ATPase p97/VCP regulates protein homeostasis using a diverse repertoire of cofactors to fulfill its biological functions. Here we use the allosteric p97 inhibitor NMS-873 to analyze its effects on enzyme composition and the ability of cells to adapt to its cytotoxicity. We found that p97 inhibition changes steady state cofactor-p97 composition, leading to the enrichment of a subset of its cofactors and polyubiquitin bound to p97. We isolated cells specifically insensitive to NMS-873 and identified a new mutation (A530T) in p97. A530T is sufficient to overcome the cytotoxicity of NMS-873 and alleviates p97 composition changes caused by the molecule but not other p97 inhibitors. This mutation does not affect NMS-873 binding but increases p97 catalytic efficiency through altered ATP and ADP binding. Collectively, these findings identify cofactor-p97 interactions sensitive to p97 inhibition and reveal a new on-target mechanism to suppress the cytotoxicity of NMS-873.


Subject(s)
Acetanilides/pharmacology , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Benzothiazoles/pharmacology , Enzyme Inhibitors/pharmacology , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Small Molecule Libraries/pharmacology , Acetanilides/chemistry , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/genetics , Allosteric Regulation/drug effects , Benzothiazoles/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , HCT116 Cells , Humans , Models, Molecular , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Small Molecule Libraries/chemistry , Structure-Activity Relationship
12.
Oncotarget ; 6(21): 18418-28, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26158899

ABSTRACT

Quiescin sulfhydryl oxidase 1 (QSOX1) is a highly conserved disulfide bond-generating enzyme that is overexpressed in diverse tumor types. Its enzymatic activity promotes the growth and invasion of tumor cells and alters extracellular matrix composition. In a nude mouse-human tumor xenograft model, tumors containing shRNA for QSOX1 grew significantly more slowly than controls, suggesting that QSOX1 supports a proliferative phenotype in vivo. High throughput screening experiments identified ebselen as an in vitro inhibitor of QSOX1 enzymatic activity. Ebselen treatment of pancreatic and renal cancer cell lines stalled tumor growth and inhibited invasion through Matrigel in vitro. Daily oral treatment with ebselen resulted in a 58% reduction in tumor growth in mice bearing human pancreatic tumor xenografts compared to controls. Mass spectrometric analysis of ebselen-treated QSOX1 mechanistically revealed that C165 and C237 of QSOX1 covalently bound to ebselen. This report details the anti-neoplastic properties of ebselen in pancreatic and renal cancer cell lines. The results here offer a "proof-of-principle" that enzymatic inhibition of QSOX1 may have clinical relevancy.


Subject(s)
Azoles/pharmacology , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Organoselenium Compounds/pharmacology , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Pancreatic Neoplasms/drug therapy , Amino Acid Sequence , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Azoles/chemistry , Blotting, Western , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cysteine/antagonists & inhibitors , Cysteine/genetics , Cysteine/metabolism , Humans , Isoindoles , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Mice, Nude , Molecular Sequence Data , Molecular Structure , Neoplasm Invasiveness , Organoselenium Compounds/chemistry , Oxidoreductases Acting on Sulfur Group Donors/antagonists & inhibitors , Oxidoreductases Acting on Sulfur Group Donors/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , RNA Interference , Sequence Homology, Amino Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
13.
ACS Comb Sci ; 17(4): 239-46, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25719760

ABSTRACT

Protein-protein interactions are generally challenging to target by small molecules. To address the challenge, we have used a multidisciplinary approach to identify small-molecule disruptors of protein-protein interactions that are mediated by SUMO (small ubiquitin-like modifier) proteins. SUMO modifications have emerged as a target with importance in treating cancer, neurodegenerative disorders, and viral infections. It has been shown that inhibiting SUMO-mediated protein-protein interactions can sensitize cancer cells to chemotherapy and radiation. We have developed highly sensitive assays using time-resolved fluorescence resonance energy transfer (TR-FRET) and fluorescence polarization (FP) that were used for high-throughput screening (HTS) to identify inhibitors for SUMO-dependent protein-protein interactions. Using these assays, we have identified a nonpeptidomimetic small molecule chemotype that binds to SUMO1 but not SUMO2 or 3. NMR chemical shift perturbation studies have shown that the compounds of this chemotype bind to the SUMO1 surface required for protein-protein interaction, despite the high sequence similarity of SUMO1 and SUMO2 and 3 at this surface.


Subject(s)
Fluorescence Resonance Energy Transfer , Small Ubiquitin-Related Modifier Proteins/chemistry , Amino Acid Motifs , Binding Sites , Ligands , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Binding , Protein Conformation
14.
J Biol Chem ; 290(12): 7693-706, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25631047

ABSTRACT

Nicotinate mononucleotide adenylyltransferase NadD is an essential enzyme in the biosynthesis of the NAD cofactor, which has been implicated as a target for developing new antimycobacterial therapies. Here we report the crystal structure of Mycobacterium tuberculosis NadD (MtNadD) at a resolution of 2.4 Å. A remarkable new feature of the MtNadD structure, compared with other members of this enzyme family, is a 310 helix that locks the active site in an over-closed conformation. As a result, MtNadD is rendered inactive as it is topologically incompatible with substrate binding and catalysis. Directed mutagenesis was also used to further dissect the structural elements that contribute to the interactions of the two MtNadD substrates, i.e. ATP and nicotinic acid mononucleotide (NaMN). For inhibitory profiling of partially active mutants and wild type MtNadD, we used a small molecule inhibitor of MtNadD with moderate affinity (Ki ∼ 25 µM) and antimycobacterial activity (MIC80) ∼ 40-80 µM). This analysis revealed interferences with some of the residues in the NaMN binding subsite consistent with the competitive inhibition observed for the NaMN substrate (but not ATP). A detailed steady-state kinetic analysis of MtNadD suggests that ATP must first bind to allow efficient NaMN binding and catalysis. This sequential mechanism is consistent with the requirement of transition to catalytically competent (open) conformation hypothesized from structural modeling. A possible physiological significance of this mechanism is to enable the down-regulation of NAD synthesis under ATP-limiting dormancy conditions. These findings point to a possible new strategy for designing inhibitors that lock the enzyme in the inactive over-closed conformation.


Subject(s)
Antitubercular Agents/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/enzymology , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Amino Acid Sequence , Antitubercular Agents/chemistry , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mycobacterium tuberculosis/drug effects , Nicotinamide-Nucleotide Adenylyltransferase/antagonists & inhibitors , Nicotinamide-Nucleotide Adenylyltransferase/chemistry , Protein Conformation , Sequence Homology, Amino Acid , Structure-Activity Relationship
15.
Biochem J ; 466(2): 311-22, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25529026

ABSTRACT

The alternative splicing of human genes is dependent on SR proteins, a family of essential splicing factors whose name derives from a signature C-terminal domain rich in arginine-serine dipeptide repeats (RS domains). Although the SRPKs (SR-specific protein kinases) phosphorylate these repeats, RS domains also contain prolines with flanking serines that are phosphorylated by a second family of protein kinases known as the CLKs (Cdc2-like kinases). The role of specific serine-proline phosphorylation within the RS domain has been difficult to assign since CLKs also phosphorylate arginine-serine dipeptides and, thus, display overlapping residue specificities with the SRPKs. In the present study, we address the effects of discrete serine-proline phosphorylation on the conformation and cellular function of the SR protein SRSF1 (SR protein splicing factor 1). Using chemical tagging and dephosphorylation experiments, we show that modification of serine-proline dipeptides broadly amplifies the conformational ensemble of SRSF1. The induction of these new structural forms triggers SRSF1 mobilization in the nucleus and alters its binding mechanism to an exonic splicing enhancer in precursor mRNA. These physical events correlate with changes in the alternative splicing of over 100 human genes based on a global splicing assay. Overall, these studies draw a direct causal relationship between a specific type of chemical modification in an SR protein and the regulation of alternative gene splicing programmes.


Subject(s)
Alternative Splicing , Nuclear Proteins/chemistry , Proline/chemistry , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , RNA Precursors/metabolism , RNA-Binding Proteins/chemistry , Amino Acid Sequence , Cell Nucleus/metabolism , Conserved Sequence , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Kinetics , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , Proline/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/genetics , Protein Transport , Protein-Tyrosine Kinases/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Serine/chemistry , Serine/metabolism , Serine-Arginine Splicing Factors , Substrate Specificity
16.
Methods ; 65(2): 165-74, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-23886911

ABSTRACT

Robust, facile high throughput assays based on non-peptidic probes are available to detect the enzyme activity of protein tyrosine phosphatases. However, these assays cannot replace the use of peptide-based probes in many applications; for example when a closer mimic of the physiological target is desired or in substrate profiling expeditions. Phosphotyrosine peptides are often used in these assays, but their use is complicated by either poor sensitivity or the need for indirect detection methods, among other pitfalls. Novel peptide-based probes for protein tyrosine phosphatases are needed to replace phosphotyrosine peptides and accelerate the field of tyrosine phosphatase substrate profiling. Here we review a type of peptidic probe for tyrosine phosphatases, which is based on the incorporation of the phosphotyrosine-mimic phosphocoumaryl amino propionic acid (pCAP) into peptides. The resulting fluorogenic pCAP peptides are dephosphorylated by tyrosine phosphatases with similar efficiency as the homologous phosphotyrosine peptides. pCAP peptides outperform phosphotyrosine peptides, providing an assay that is as robust, sensitive and facile as the non-peptidic fluorogenic probes on the market. Finally the use of pCAP can expand the range of phosphatase assays, facilitating the investigation of multiphosphorylated peptides and providing an in-gel assay for phosphatase activity.


Subject(s)
Alanine/analogs & derivatives , Biological Assay/methods , Coumarins/chemistry , Fluorescent Dyes/chemistry , Organophosphates/chemistry , Peptides/chemistry , Protein Tyrosine Phosphatases/chemistry , Alanine/chemistry , Electrophoresis, Polyacrylamide Gel , Humans , Peptides/genetics , Protein Tyrosine Phosphatases/genetics , Substrate Specificity
17.
Methods Mol Biol ; 1053: 103-13, 2013.
Article in English | MEDLINE | ID: mdl-23860649

ABSTRACT

Tissue nonspecific alkaline phosphatase (TNAP) is one of four human alkaline phosphatases (AP), a family of exocytic enzymes that catalyze hydrolysis of phospho-monoesters in bone, liver, kidney, and various other tissues. Overexpression of TNAP gives rise to excessive bone and soft tissue mineralization, including blood vessel calcification. Our prior screening campaigns have found several leads against this attractive therapeutic target using in vitro assay with a recombinant enzyme; these compounds were further optimized using medicinal chemistry approaches. To prioritize compounds for their use in animal models, we have designed and developed a biomarker assay for in situ detection of TNAP activity within human and mouse blood samples at physiological pH. This assay is suitable for screening compounds in 1,536-well plates using blood plasma from different mammalian species. The user may choose from two different substrates based on the need for greater assay simplicity or sensitivity.


Subject(s)
Alkaline Phosphatase/antagonists & inhibitors , Blood Chemical Analysis/methods , Enzyme Assays , Enzyme Inhibitors/blood , High-Throughput Screening Assays , Alkaline Phosphatase/blood , Animals , Biomarkers/blood , Calcification, Physiologic , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Mice
18.
Methods Mol Biol ; 1053: 145-54, 2013.
Article in English | MEDLINE | ID: mdl-23860653

ABSTRACT

Existing assays monitoring ENPP1 activity are either not physiologically relevant or not suitable for high-throughput screening (HTS). Here, we describe the development and implementation of two new ENPP1 activity assays that address these drawbacks. These assays employ physiological substrates of ENPP1, ATP and ADP. They rely on detection of inorganic phosphate using a special modification of the malachite green-molybdate colorimetric procedure that ensures stability of acid-labile compounds, such as the ones containing phosphodiester bonds. The pyrophosphate generated in ENPP1 reaction is converted to inorganic phosphate in the presence of inorganic phosphatase; whereas, omission of this coupling enzyme enables detection of the inorganic phosphate generated by ENPP1. These new ENPP1 assays were miniaturized into high-density microplate formats. With minimal requirement for ENPP1 enzyme, low micromolar phosphate detection sensitivity, and simple protocol involving three to four simple liquid handling steps, these robust assays are suitable for HTS.


Subject(s)
Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Enzyme Assays , High-Throughput Screening Assays , Phosphoric Diester Hydrolases/analysis , Pyrophosphatases/analysis , Cell Line , Chondrocytes/enzymology , HEK293 Cells , Humans , Osteoblasts/enzymology , Phosphates/analysis , Phosphoric Diester Hydrolases/chemistry , Pyrophosphatases/chemistry
19.
J Mol Biol ; 425(16): 2894-909, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23707382

ABSTRACT

SR proteins are essential splicing factors whose biological function is regulated through phosphorylation of their C-terminal RS domains. Prior studies have shown that cytoplasmic-nuclear translocalization of the SR protein SRSF1 is regulated by multisite phosphorylation of a long Arg-Ser repeat in the N-terminus of the RS domain while subnuclear localization is controlled by phosphorylation of a shorter Arg-Ser repeat along with several Ser-Pro dipeptides in the C-terminus of the RS domain. To better understand how these two kinases partition Arg-Ser versus Ser-Pro specificities, we monitored the phosphorylation of SRSF1 by CLK1 and SRPK1. Although SRPK1 initially binds at the center of the RS domain phosphorylating in an orderly, N-terminal direction, CLK1 makes widespread contacts in the RS domain and generates multiple enzyme-substrate complexes that induce a random addition mechanism. While SRPK1 rapidly phosphorylates N-terminal serines, SRPK1 and CLK1 display similar activities toward Arg-Ser repeats in the C-terminus, suggesting that these kinases may not separate function in a strict linear manner along the RS domain. CLK1 induces a unique gel shift in SRSF1 that is not the result of enhanced Arg-Ser phosphorylation but rather is the direct result of the phosphorylation of several Ser-Pro dipeptides. These prolines are important for binding and phosphorylation of the SR protein by CLK1 but not for the SRPK1-dependent reaction. The data establish a new view of SR protein regulation in which SRPK1 and CLK1 partition activities based on Ser-Pro versus Arg-Ser placement rather than on N- and C-terminal preferences along the RS domain.


Subject(s)
Nuclear Proteins/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , RNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay , Humans , Models, Biological , Phosphorylation , Serine-Arginine Splicing Factors
20.
J Mol Biol ; 410(1): 131-45, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21600902

ABSTRACT

SR proteins (splicing factors containing arginine-serine repeats) are essential splicing factors whose phosphorylation by the SR-specific protein kinase (SRPK) family regulates nuclear localization and mRNA processing activity. In addition to an N-terminal extension with unknown function, SRPKs contain a large, nonhomologous spacer insert domain (SID) that bifurcates the kinase domain and anchors the kinase in the cytoplasm through interactions with chaperones. While structures for the kinase domain are now available, constructs that include regions outside this domain have been resistant to crystallographic elucidation. To investigate the conformation of the full-length kinase and the functional role of noncatalytic regions, we performed hydrogen-deuterium exchange and steady-state kinetic experiments on SRPK1. Unlike the kinase core, the large SID lacks stable, hydrogen-bonded structure and may provide an intrinsically disordered region for chaperone interactions. Conversely, the N-terminus, which positively regulates SR protein binding, adopts a stable structure when the insert domain is present and stabilizes a docking groove in the large lobe of the kinase domain. The N-terminus and SID equally enhance SR protein turnover by altering the stability of several catalytic loop segments. These studies reveal that SRPK1 uses an N-terminal extension and a large, intrinsically disordered region juxtaposed to a stable structure to facilitate high-affinity SR protein interactions and phosphorylation rates.


Subject(s)
Deuterium Exchange Measurement , Gene Expression Regulation , Molecular Chaperones/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , HeLa Cells , Humans , Models, Molecular , Mutagenesis , Phosphorylation , Protein Conformation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Structure, Tertiary , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...