Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
RNA ; 29(9): 1400-1410, 2023 09.
Article in English | MEDLINE | ID: mdl-37279998

ABSTRACT

Unique chemical and physical properties are introduced by inserting selenocysteine (Sec) at specific sites within proteins. Recombinant and facile production of eukaryotic selenoproteins would benefit from a yeast expression system; however, the selenoprotein biosynthetic pathway was lost in the evolution of the kingdom Fungi as it diverged from its eukaryotic relatives. Based on our previous development of efficient selenoprotein production in bacteria, we designed a novel Sec biosynthesis pathway in Saccharomyces cerevisiae using Aeromonas salmonicida translation components. S. cerevisiae tRNASer was mutated to resemble A. salmonicida tRNASec to allow recognition by S. cerevisiae seryl-tRNA synthetase as well as A. salmonicida selenocysteine synthase (SelA) and selenophosphate synthetase (SelD). Expression of these Sec pathway components was then combined with metabolic engineering of yeast to enable the production of active methionine sulfate reductase enzyme containing genetically encoded Sec. Our report is the first demonstration that yeast is capable of selenoprotein production by site-specific incorporation of Sec.


Subject(s)
Saccharomyces cerevisiae , Codon, Terminator/genetics , Codon, Terminator/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Aeromonas salmonicida/genetics , Protein Engineering , RNA, Transfer, Cys/chemistry , RNA, Transfer, Cys/genetics , RNA, Transfer, Cys/metabolism , Humans , Nucleic Acid Conformation
2.
Cell Mol Life Sci ; 78(10): 4589-4613, 2021 May.
Article in English | MEDLINE | ID: mdl-33855626

ABSTRACT

Since its discovery in 1981, the Ku complex has been extensively studied under multiple cellular contexts, with most work focusing on Ku in terms of its essential role in non-homologous end-joining (NHEJ). In this process, Ku is well-known as the DNA-binding subunit for DNA-PK, which is central to the NHEJ repair process. However, in addition to the extensive study of Ku's role in DNA repair, Ku has also been implicated in various other cellular processes including transcription, the DNA damage response, DNA replication, telomere maintenance, and has since been studied in multiple contexts, growing into a multidisciplinary point of research across various fields. Some advances have been driven by clarification of Ku's structure, including the original Ku crystal structure and the more recent Ku-DNA-PKcs crystallography, cryogenic electron microscopy (cryoEM) studies, and the identification of various post-translational modifications. Here, we focus on the advances made in understanding the Ku heterodimer outside of non-homologous end-joining, and across a variety of model organisms. We explore unique structural and functional aspects, detail Ku expression, conservation, and essentiality in different species, discuss the evidence for its involvement in a diverse range of cellular functions, highlight Ku protein interactions and recent work concerning Ku-binding motifs, and finally, we summarize the clinical Ku-related research to date.


Subject(s)
DNA End-Joining Repair/genetics , Ku Autoantigen/genetics , Animals , DNA-Binding Proteins/genetics , Humans , Protein Processing, Post-Translational/genetics , Telomere/genetics
3.
J Biol Chem ; 295(24): 8120-8134, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32350110

ABSTRACT

Protein kinase B (AKT1) is a central node in a signaling pathway that regulates cell survival. The diverse pathways regulated by AKT1 are communicated in the cell via the phosphorylation of perhaps more than 100 cellular substrates. AKT1 is itself activated by phosphorylation at Thr-308 and Ser-473. Despite the fact that these phosphorylation sites are biomarkers for cancers and tumor biology, their individual roles in shaping AKT1 substrate selectivity are unknown. We recently developed a method to produce AKT1 with programmed phosphorylation at either or both of its key regulatory sites. Here, we used both defined and randomized peptide libraries to map the substrate selectivity of site-specific, singly and doubly phosphorylated AKT1 variants. To globally quantitate AKT1 substrate preferences, we synthesized three AKT1 substrate peptide libraries: one based on 84 "known" substrates and two independent and larger oriented peptide array libraries (OPALs) of ∼1011 peptides each. We found that each phospho-form of AKT1 has common and distinct substrate requirements. Compared with pAKT1T308, the addition of Ser-473 phosphorylation increased AKT1 activities on some, but not all of its substrates. This is the first report that Ser-473 phosphorylation can positively or negatively regulate kinase activity in a substrate-dependent fashion. Bioinformatics analysis indicated that the OPAL-activity data effectively discriminate known AKT1 substrates from closely related kinase substrates. Our results also enabled predictions of novel AKT1 substrates that suggest new and expanded roles for AKT1 signaling in regulating cellular processes.


Subject(s)
Proto-Oncogene Proteins c-akt/metabolism , Amino Acid Motifs , Amino Acid Sequence , Humans , Peptide Library , Peptides/chemistry , Peptides/metabolism , Phosphorylation , Phosphoserine/metabolism , Proto-Oncogene Proteins c-akt/chemistry , ROC Curve , Substrate Specificity
4.
RNA Biol ; 18(sup1): 397-408, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34288801

ABSTRACT

In the cell, RNA abundance is dynamically controlled by transcription and decay rates. Posttranscriptional nucleotide addition at the RNA 3' end is a means of regulating mRNA and RNA stability and activity, as well as marking RNAs for degradation. The human nucleotidyltransferase Gld2 polyadenylates mRNAs and monoadenylates microRNAs, leading to an increase in RNA stability. The broad substrate range of Gld2 and its role in controlling RNA stability make the regulation of Gld2 activity itself imperative. Gld2 activity can be regulated by post-translational phosphorylation via the oncogenic kinase Akt1 and other kinases, leading to either increased or almost abolished enzymatic activity, and here we confirm that Akt1 phosphorylates Gld2 in a cellular context. Another means to control Gld2 RNA specificity and activity is the interaction with RNA binding proteins. Known interactors are QKI-7 and CPEB, which recruit Gld2 to specific miRNAs and mRNAs. We investigate the interplay between five phosphorylation sites in the N-terminal domain of Gld2 and three RNA binding proteins. We found that the activity and RNA specificity of Gld2 is dynamically regulated by this network. Binding of QKI-7 or phosphorylation at S62 relieves the autoinhibitory function of the Gld2 N-terminal domain. Binding of QKI-7 to a short peptide sequence within the N-terminal domain can also override the deactivation caused by Akt1 phosphorylation at S116. Our data revealed that Gld2 substrate specificity and activity can be dynamically regulated to match the cellular need of RNA stabilization and turnover.


Subject(s)
Adenine/chemistry , Gene Expression Regulation , MicroRNAs/metabolism , Polynucleotide Adenylyltransferase/metabolism , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Adenine/metabolism , HEK293 Cells , Humans , MicroRNAs/genetics , Phosphorylation , Polynucleotide Adenylyltransferase/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Substrate Specificity , mRNA Cleavage and Polyadenylation Factors/genetics
5.
J Biol Chem ; 293(27): 10744-10756, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29773654

ABSTRACT

The proto-oncogene Akt/protein kinase B (PKB) is a pivotal signal transducer for growth and survival. Growth factor stimulation leads to Akt phosphorylation at two regulatory sites (Thr-308 and Ser-473), acutely activating Akt signaling. Delineating the exact role of each regulatory site is, however, technically challenging and has remained elusive. Here, we used genetic code expansion to produce site-specifically phosphorylated Akt1 to dissect the contribution of each regulatory site to Akt1 activity. We achieved recombinant production of full-length Akt1 containing site-specific pThr and pSer residues for the first time. Our analysis of Akt1 site-specifically phosphorylated at either or both sites revealed that phosphorylation at both sites increases the apparent catalytic rate 1500-fold relative to unphosphorylated Akt1, an increase attributable primarily to phosphorylation at Thr-308. Live imaging of COS-7 cells confirmed that phosphorylation of Thr-308, but not Ser-473, is required for cellular activation of Akt. We found in vitro and in the cell that pThr-308 function cannot be mimicked with acidic residues, nor could unphosphorylated Thr-308 be mimicked by an Ala mutation. An Akt1 variant with pSer-308 achieved only partial enzymatic and cellular signaling activity, revealing a critical interaction between the γ-methyl group of pThr-308 and Cys-310 in the Akt1 active site. Thus, pThr-308 is necessary and sufficient to stimulate Akt signaling in cells, and the common use of phosphomimetics is not appropriate for studying the biology of Akt signaling. Our data also indicate that pThr-308 should be regarded as the primary diagnostic marker of Akt activity.


Subject(s)
Genetic Code , Molecular Imaging/methods , Proto-Oncogene Proteins c-akt/metabolism , Serine/metabolism , Threonine/metabolism , Cells, Cultured , Crystallography, X-Ray , Humans , Mutation , Phosphorylation , Protein Conformation , Proto-Oncogene Mas , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/genetics , Serine/chemistry , Serine/genetics , Threonine/chemistry , Threonine/genetics
6.
RNA Biol ; 16(8): 1022-1033, 2019 08.
Article in English | MEDLINE | ID: mdl-31057087

ABSTRACT

The de-regulation of microRNAs (miRNAs) is associated with multiple human diseases, yet cellular mechanisms governing miRNA abundance remain largely elusive. Human miR-122 is required for Hepatitis C proliferation, and low miR-122 abundance is associated with hepatic cancer. The adenylyltransferase Gld2 catalyses the post-transcriptional addition of a single adenine residue (A + 1) to the 3'-end of miR-122, enhancing its stability. Gld2 activity is inhibited by binding to the Hepatitis C virus core protein during HepC infection, but no other mechanisms of Gld2 regulation are known. We found that Gld2 activity is regulated by site-specific phosphorylation in its disordered N-terminal domain. We identified two phosphorylation sites (S62, S110) where phosphomimetic substitutions increased Gld2 activity and one site (S116) that markedly reduced activity. Using mass spectrometry, we confirmed that HEK 293 cells readily phosphorylate the N-terminus of Gld2. We identified protein kinase A (PKA) and protein kinase B (Akt1) as the kinases that site-specifically phosphorylate Gld2 at S116, abolishing Gld2-mediated nucleotide addition. The data demonstrate a novel phosphorylation-dependent mechanism to regulate Gld2 activity, revealing tumour suppressor miRNAs as a previously unknown target of Akt1-dependent signalling.


Subject(s)
Liver Neoplasms/genetics , MicroRNAs/genetics , Polynucleotide Adenylyltransferase/genetics , Proto-Oncogene Proteins c-akt/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , Cell Proliferation/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , HEK293 Cells , Hepatitis C/genetics , Hepatitis C/pathology , Hepatitis C/virology , Humans , Liver Neoplasms/pathology , Liver Neoplasms/virology , Phosphorylation , Protein Domains/genetics , Signal Transduction/genetics
7.
Biochim Biophys Acta Mol Cell Res ; 1871(7): 119815, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39151475

ABSTRACT

The Ku heterodimer (Ku70/Ku80) is central to the non-homologous end-joining (NHEJ) pathway. Ku binds to the broken DNA ends and promotes the assembly of the DNA repair complex. The N-terminal Ku70 von Willebrand A (vWA) domain is known to mediate protein-protein interactions important for the repair process. In particular, the D192 and D195 residues within helix 5 of the Ku70 vWA domain were shown to be essential for NHEJ function, although the precise role of these residues was not identified. Here, we set up a miniTurbo screening system to identify Ku70 D192/D195 residue-specific interactors in a conditional, human Ku70-knockout cell line in response to DNA damage. Using fusion protein constructs of Ku70 wild-type and mutant (D192A/D195R) with miniTurbo, we identified a number of candidate proximal interactors in response to DNA damage treatment, including DNA Ligase IV (LigIV), a known and essential NHEJ complex member. Interestingly, LigIV was enriched in our wildtype screen but not the Ku70 D192A/D195R screen, suggesting its interaction is disrupted by the mutation. Validation experiments demonstrated that the DNA damage-induced interaction between Ku70 and LigIV was disrupted by the Ku70 D192A/D195R mutations. Our findings provide greater detail about the interaction surface between the Ku70 vWA domain and LigIV and offer strong evidence that the D192 and D195 residues are important for NHEJ completion through an interaction with LigIV. Altogether, this work reveals novel potential proximal interactors of Ku in response to DNA damage and identifies Ku70 D192/D195 residues as essential for LigIV interaction with Ku during NHEJ.


Subject(s)
DNA End-Joining Repair , DNA Ligase ATP , DNA-Binding Proteins , Ku Autoantigen , Ku Autoantigen/metabolism , Ku Autoantigen/genetics , Humans , DNA End-Joining Repair/genetics , DNA Ligase ATP/metabolism , DNA Ligase ATP/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/chemistry , DNA Damage , Protein Binding , Protein Domains , von Willebrand Factor/metabolism , von Willebrand Factor/genetics , von Willebrand Factor/chemistry
8.
Cells ; 11(23)2022 Nov 29.
Article in English | MEDLINE | ID: mdl-36497091

ABSTRACT

Protein kinase B (AKT1) is a serine/threonine kinase and central transducer of cell survival pathways. Typical approaches to study AKT1 biology in cells rely on growth factor or insulin stimulation that activates AKT1 via phosphorylation at two key regulatory sites (Thr308, Ser473), yet cell stimulation also activates many other kinases. To produce cells with specific AKT1 activity, we developed a novel system to deliver active AKT1 to human cells. We recently established a method to produce AKT1 phospho-variants from Escherichia coli with programmed phosphorylation. Here, we fused AKT1 with an N-terminal cell penetrating peptide tag derived from the human immunodeficiency virus trans-activator of transcription (TAT) protein. The TAT-tag did not alter AKT1 kinase activity and was necessary and sufficient to rapidly deliver AKT1 protein variants that persisted in human cells for 24 h without the need to use transfection reagents. TAT-pAKT1T308 induced selective phosphorylation of the known AKT1 substrate GSK-3α, but not GSK-3ß, and downstream stimulation of the AKT1 pathway as evidenced by phosphorylation of ribosomal protein S6 at Ser240/244. The data demonstrate efficient delivery of AKT1 with programmed phosphorylation to human cells, thus establishing a cell-based model system to investigate signaling that is dependent on AKT1 activity.


Subject(s)
Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-akt , Humans , Proto-Oncogene Proteins c-akt/metabolism , Protein Serine-Threonine Kinases/metabolism , Phosphorylation , Signal Transduction , Insulin/metabolism
9.
Cells ; 11(5)2022 02 26.
Article in English | MEDLINE | ID: mdl-35269443

ABSTRACT

The phosphoinositide-3-kinase (PI3K)/AKT pathway regulates cell survival and is over-activated in most human cancers, including ovarian cancer. Following growth factor stimulation, AKT1 is activated by phosphorylation at T308 and S473. Disruption of the AKT1 signaling pathway is sufficient to inhibit the epithelial-mesenchymal transition in epithelial ovarian cancer (EOC) cells. In metastatic disease, adherent EOC cells transition to a dormant spheroid state, characterized previously by low S473 phosphorylation in AKT1. We confirmed this finding and observed that T308 phosphorylation was yet further reduced in EOC spheroids and that the transition from adherent to spheroid growth is accompanied by significantly increased levels of let-7 miRNAs. We then used mechanistic studies to investigate the impact of let-7 miRNAs on AKT1 phosphorylation status and activity in cells. In growth factor-stimulated HEK 293T cells supplemented with let-7a, we found increased phosphorylation of AKT1 at T308, decreased phosphorylation at S473, and enhanced downstream AKT1 substrate GSK-3ß phosphorylation. Let-7b and let-7g also deregulated AKT signaling by rendering AKT1 insensitive to growth factor simulation. We uncovered let-7a-dependent deregulation of PI3K pathway components, including PI3KC2A, PDK1, and RICTOR, that govern AKT1 phosphorylation and activity. Together, our data show a new role for miRNAs in regulating AKT signaling.


Subject(s)
MicroRNAs , Ovarian Neoplasms , Carcinoma, Ovarian Epithelial , Glycogen Synthase Kinase 3 beta/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
10.
Front Bioeng Biotechnol ; 8: 619252, 2020.
Article in English | MEDLINE | ID: mdl-33614606

ABSTRACT

Protein kinase B (AKT1) is hyper-activated in diverse human tumors. AKT1 is activated by phosphorylation at two key regulatory sites, Thr308 and Ser473. Active AKT1 phosphorylates many, perhaps hundreds, of downstream cellular targets in the cytosol and nucleus. AKT1 is well-known for phosphorylating proteins that regulate cell survival and apoptosis, however, the full catalog of AKT1 substrates remains unknown. Using peptide arrays, we recently discovered that each phosphorylated form of AKT1 (pAKT1S473, pAKT1T308, and ppAKT1S473,T308) has a distinct substrate specificity, and these data were used to predict potential new AKT1 substrates. To test the high-confidence predictions, we synthesized target peptides representing putative AKT1 substrates. Peptides substrates were synthesized by solid phase synthesis and their purity was confirmed by mass spectrometry. Most of the predicted peptides showed phosphate accepting activity similar to or greater than that observed with a peptide derived from a well-established AKT1 substrate, glycogen synthase kinase 3ß (GSK-3ß). Among the novel substrates, AKT1 was most active with peptides representing PIP3-binding protein Rab11 family-interacting protein 2 and cysteinyl leukotriene receptor 1, indicating their potential role in AKT1-dependent cellular signaling. The ppAKT1S473,T308 enzyme was highly selective for peptides containing a patch of basic residues at -5, -4, -3 and aromatic residues (Phe/Tyr) at +1 positions from the phosphorylation site. The pAKT1S473 variant preferred more acidic peptides, Ser or Pro at +4, and was agnostic to the residue at -5. The data further support our hypothesis that Ser473 phosphorylation plays a key role in modulating AKT1 substrate selectivity.

11.
Genes (Basel) ; 9(9)2018 Sep 07.
Article in English | MEDLINE | ID: mdl-30205513

ABSTRACT

Protein kinase B (Akt1) is a proto-oncogene that is overactive in most cancers. Akt1 activation requires phosphorylation at Thr308; phosphorylation at Ser473 further enhances catalytic activity. Akt1 activity is also regulated via interactions between the kinase domain and the N-terminal auto-inhibitory pleckstrin homology (PH) domain. As it was previously difficult to produce Akt1 in site-specific phosphorylated forms, the contribution of each activating phosphorylation site to auto-inhibition was unknown. Using a combination of genetic code expansion and in vivo enzymatic phosphorylation, we produced Akt1 variants containing programmed phosphorylation to probe the interplay between Akt1 phosphorylation status and the auto-inhibitory function of the PH domain. Deletion of the PH domain increased the enzyme activity for all three phosphorylated Akt1 variants. For the doubly phosphorylated enzyme, deletion of the PH domain relieved auto-inhibition by 295-fold. We next found that phosphorylation at Ser473 provided resistance to chemical inhibition by Akti-1/2 inhibitor VIII. The Akti-1/2 inhibitor was most effective against pAkt1T308 and showed four-fold decreased potency with Akt1 variants phosphorylated at Ser473. The data highlight the need to design more potent Akt1 inhibitors that are effective against the doubly phosphorylated and most pathogenic form of Akt1.

12.
Food Chem ; 135(4): 2320-5, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-22980808

ABSTRACT

Hypertension is a major public health problem rising across the globe. Inhibition of angiotensin converting enzyme (ACE) is identified as a main therapeutic target in controlling high blood pressure. The present study investigated the ACE inhibitory property of a flavonoid-rich apple peel extract (FAE), its constituents, selected flavonoids and some quercetin metabolites using a biochemical assay of ACE inhibition and a human umbilical vein endothelial cell (HUVEC) model. FAE, all the tested flavonoids except genistein, and two quercetin metabolites (quercetin-3-O-glucuronic acid and quercetin-3-O-sulfate) significantly (p<0.05) inhibited ACE. Enzyme kinetic analysis revealed that flavonoids are competitive inhibitors of ACE. In the HUVEC model, FAE, quercetin-3-O-glucoside and quercetin-3-O-glucuronic acid inhibited significantly (p<0.05) ACE activity. Overall, FAE and most of the flavonoids tested showed ACE inhibition in vitro which needs further investigations using animal and human clinical trials.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/chemistry , Antihypertensive Agents/chemistry , Flavonoids/chemistry , Malus/chemistry , Plant Extracts/chemistry , Animals , Fruit/chemistry , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Kinetics , Peptidyl-Dipeptidase A/chemistry , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL