Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38260389

ABSTRACT

The C-terminal domain of RPB1 (CTD) orchestrates transcription by recruiting regulators to RNA Pol II upon phosphorylation. Recent insights highlight the pivotal role of CTD in driving condensate formation on gene loci. Yet, the molecular mechanism behind how CTD-mediated recruitment of transcriptional regulators influences condensates formation remains unclear. Our study unveils that phosphorylation reversibly dissolves phase separation induced by the unphosphorylated CTD. Phosphorylated CTD, upon specific association with transcription regulatory proteins, forms distinct condensates from unphosphorylated CTD. Function studies demonstrate CTD variants with diverse condensation properties in vitro exhibit difference in promoter binding and mRNA co-processing in cells. Notably, varying CTD lengths lead to alternative splicing outcomes impacting cellular growth, linking the evolution of CTD variation/length with the complexity of splicing from yeast to human. These findings provide compelling evidence for a model wherein post-translational modification enables the transition of functionally specialized condensates, highlighting a co-evolution link between CTD condensation and splicing.

2.
Cell Metab ; 35(6): 1009-1021.e9, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37084733

ABSTRACT

Insulin inhibits gluconeogenesis and stimulates glucose conversion to glycogen and lipids. How these activities are coordinated to prevent hypoglycemia and hepatosteatosis is unclear. Fructose-1,6-bisphosphatase (FBP1) is rate controlling for gluconeogenesis. However, inborn human FBP1 deficiency does not cause hypoglycemia unless accompanied by fasting or starvation, which also trigger paradoxical hepatomegaly, hepatosteatosis, and hyperlipidemia. Hepatocyte FBP1-ablated mice exhibit identical fasting-conditional pathologies along with AKT hyperactivation, whose inhibition reversed hepatomegaly, hepatosteatosis, and hyperlipidemia but not hypoglycemia. Surprisingly, fasting-mediated AKT hyperactivation is insulin dependent. Independently of its catalytic activity, FBP1 prevents insulin hyperresponsiveness by forming a stable complex with AKT, PP2A-C, and aldolase B (ALDOB), which specifically accelerates AKT dephosphorylation. Enhanced by fasting and weakened by elevated insulin, FBP1:PP2A-C:ALDOB:AKT complex formation, which is disrupted by human FBP1 deficiency mutations or a C-terminal FBP1 truncation, prevents insulin-triggered liver pathologies and maintains lipid and glucose homeostasis. Conversely, an FBP1-derived complex disrupting peptide reverses diet-induced insulin resistance.


Subject(s)
Fructose , Hypoglycemia , Humans , Mice , Animals , Fructose-Bisphosphatase/genetics , Proto-Oncogene Proteins c-akt , Insulin , Hepatomegaly/complications , Hypoglycemia/etiology , Glucose
3.
Curr Opin Chem Biol ; 74: 102279, 2023 06.
Article in English | MEDLINE | ID: mdl-36966700

ABSTRACT

Eukaryotes depend upon the proper localization, accumulation, and release of intracellular Ca2+. This is regulated through specialized cellular compartments, signaling pathways, and Ca2+-binding proteins and channels. Cytosolic and extracellular signaling governing intracellular Ca2+ stores are well explored. However, regulatory signals within Ca2+ storage organelles like the endoplasmic/sarcoplasmic reticulum are not well understood. This is due to a lack of identified signaling molecules - like protein kinases - within these compartments, limited information on their regulation, and incomplete understanding of mechanisms involving modified substrates. Here we review recent advances in intralumenal signaling focusing on the secretory pathway protein kinase FAM20C and its regulation, Ca2+-binding protein substrates, and potential mechanisms through which FAM20C may regulate Ca2+ storage.


Subject(s)
Calcium , Sarcoplasmic Reticulum , Sarcoplasmic Reticulum/metabolism , Calcium/metabolism , Secretory Pathway , Endoplasmic Reticulum/metabolism , Phosphotransferases/metabolism
4.
J Bone Miner Res ; 36(8): 1548-1565, 2021 08.
Article in English | MEDLINE | ID: mdl-33905568

ABSTRACT

Proper embryonic and postnatal skeletal development require coordination of myriad complex molecular mechanisms. Disruption of these processes, through genetic mutation, contributes to variation in skeletal development. We developed a high-throughput N-ethyl-N-nitrosourea (ENU)-induced saturation mutagenesis skeletal screening approach in mice to identify genes required for proper skeletal development. Here, we report initial results from live-animal X-ray and dual-energy X-ray absorptiometry (DXA) imaging of 27,607 G3 mice from 806 pedigrees, testing the effects of 32,198 coding/splicing mutations in 13,020 genes. A total of 39.7% of all autosomal genes were severely damaged or destroyed by mutations tested twice or more in the homozygous state. Results from our study demonstrate the feasibility of in vivo mutagenesis to identify mouse models of skeletal disease. Furthermore, our study demonstrates how ENU mutagenesis provides opportunities to create and characterize putative hypomorphic mutations in developmentally essential genes. Finally, we present a viable mouse model and case report of recessive skeletal disease caused by mutations in FAM20B. Results from this study, including engineered mouse models, are made publicly available via the online Mutagenetix database. © 2021 American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Bone Diseases/genetics , Germ Cells , Mutagenesis , Animals , Ethylnitrosourea , Humans , Mice , Mutation , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/genetics
5.
J Biol Chem ; 296: 100267, 2021.
Article in English | MEDLINE | ID: mdl-33759783

ABSTRACT

The study of extracellular phosphorylation was initiated in late 19th century when the secreted milk protein, casein, and egg-yolk protein, phosvitin, were shown to be phosphorylated. However, it took more than a century to identify Fam20C, which phosphorylates both casein and phosvitin under physiological conditions. This kinase, along with its family members Fam20A and Fam20B, defined a new family with altered amino acid sequences highly atypical from the canonical 540 kinases comprising the kinome. Fam20B is a glycan kinase that phosphorylates xylose residues and triggers peptidoglycan biosynthesis, a role conserved from sponges to human. The protein kinase, Fam20C, conserved from nematodes to humans, phosphorylates well over 100 substrates in the secretory pathway with overall functions postulated to encompass endoplasmic reticulum homeostasis, nutrition, cardiac function, coagulation, and biomineralization. The preferred phosphorylation motif of Fam20C is SxE/pS, and structural studies revealed that related member Fam20A allosterically activates Fam20C by forming a heterodimeric/tetrameric complex. Fam20A, a pseudokinase, is observed only in vertebrates. Loss-of-function genetic alterations in the Fam20 family lead to human diseases such as amelogenesis imperfecta, nephrocalcinosis, lethal and nonlethal forms of Raine syndrome with major skeletal defects, and altered phosphate homeostasis. Together, these three members of the Fam20 family modulate a diverse network of secretory pathway components playing crucial roles in health and disease. The overarching theme of this review is to highlight the progress that has been made in the emerging field of extracellular phosphorylation and the key roles secretory pathway kinases play in an ever-expanding number of cellular processes.


Subject(s)
Casein Kinase I/metabolism , Extracellular Matrix Proteins/metabolism , Casein Kinase I/chemistry , Endoplasmic Reticulum/metabolism , Extracellular Matrix Proteins/chemistry , Homeostasis , Humans , Myocardium/metabolism , Phosphorylation , Secretory Pathway , Signal Transduction , Structure-Activity Relationship , Substrate Specificity
6.
J Biol Chem ; 296: 100184, 2021.
Article in English | MEDLINE | ID: mdl-33310704

ABSTRACT

Magnesium ions play a critical role in catalysis by many enzymes and contribute to the fidelity of DNA polymerases through a two-metal ion mechanism. However, specificity is a kinetic phenomenon and the roles of Mg2+ ions in each step in the catalysis have not been resolved. We first examined the roles of Mg2+ by kinetic analysis of single nucleotide incorporation catalyzed by HIV reverse transcriptase. We show that Mg.dNTP binding induces an enzyme conformational change at a rate that is independent of free Mg2+ concentration. Subsequently, the second Mg2+ binds to the closed state of the enzyme-DNA-Mg.dNTP complex (Kd = 3.7 mM) to facilitate catalysis. Weak binding of the catalytic Mg2+ contributes to fidelity by sampling the correctly aligned substrate without perturbing the equilibrium for nucleotide binding at physiological Mg2+ concentrations. An increase of the Mg2+ concentration from 0.25 to 10 mM increases nucleotide specificity (kcat/Km) 12-fold largely by increasing the rate of the chemistry relative to the rate of nucleotide release. Mg2+ binds very weakly (Kd ≤ 37 mM) to the open state of the enzyme. Analysis of published crystal structures showed that HIV reverse transcriptase binds only two metal ions prior to incorporation of a correct base pair. Molecular dynamics simulations support the two-metal ion mechanism and the kinetic data indicating weak binding of the catalytic Mg2+. Molecular dynamics simulations also revealed the importance of the divalent cation cloud surrounding exposed phosphates on the DNA. These results enlighten the roles of the two metal ions in the specificity of DNA polymerases.


Subject(s)
HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , Magnesium/metabolism , Cations, Divalent/chemistry , Cations, Divalent/metabolism , HIV Infections/virology , HIV Reverse Transcriptase/chemistry , HIV-1/chemistry , HIV-1/metabolism , Humans , Kinetics , Magnesium/chemistry , Molecular Dynamics Simulation , Protein Conformation , Thermodynamics
7.
ACS Chem Biol ; 15(8): 2259-2272, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32568517

ABSTRACT

The phosphorylation states of RNA polymerase II coordinate the process of eukaryotic transcription by recruitment of transcription regulators. The individual residues of the repetitive heptad of the C-terminal domain (CTD) of the biggest subunit of RNA polymerase II are phosphorylated temporally at different stages of transcription. Intriguingly, despite similar flanking residues, phosphorylation of Ser2 and Ser5 in CTD heptads play dramatically different roles. The mechanism of how the kinases place phosphorylation on the correct serine is not well understood. In this paper, we use biochemical assays, mass spectrometry, molecular modeling, and structural analysis to understand the structural elements determining which serine of the CTD heptad is subject to phosphorylation. We identified three motifs in the activation/P+1 loops differentiating the intrinsic specificity of CTD in various CTD kinases. We characterized the enzyme specificity of the CTD kinases-CDK7 as Ser5-specific, Erk2 with dual specificity for Ser2 and Ser5, and Dyrk1a as a Ser2-specific kinase. We also show that the specificities of kinases are malleable and can be modified by incorporating mutations in their activation/P+1 loops that alter the interactions of the three motifs. Our results provide an important clue to the understanding of post-translational modification of RNA polymerase II temporally during active transcription.


Subject(s)
Protein Kinases/metabolism , RNA Polymerase II/metabolism , Transcription, Genetic , Amino Acid Sequence , Cyclin-Dependent Kinases/metabolism , Discoidin Domain Receptor 1 , Humans , Mass Spectrometry/methods , Phosphorylation , Protein Kinases/chemistry , Protein Processing, Post-Translational , RNA Polymerase II/chemistry , Sequence Homology, Amino Acid , Substrate Specificity , Cyclin-Dependent Kinase-Activating Kinase
8.
Bio Protoc ; 10(12): e3648, 2020 Jun 20.
Article in English | MEDLINE | ID: mdl-33659319

ABSTRACT

Eukaryotic RNA polymerase II transcribes all protein-coding mRNAs and is highly regulated. A key mechanism directing RNA polymerase II and facilitating the co-transcriptional processing of mRNAs is the phosphorylation of its highly repetitive carboxyl-terminal domain (CTD) of its largest subunit, RPB1, at specific residues. A variety of techniques exist to identify and quantify the degree of CTD phosphorylation, including phosphorylation-specific antibodies and mass spectrometry. Electrophoretic mobility shift assays (EMSAs) have been utilized since the discovery of CTD phosphorylation and continue to represent a simple, direct, and widely applicable approach for qualitatively monitoring CTD phosphorylation. We present a standardized method for EMSA analysis of recombinant GST-CTD substrates phosphorylated by a variety of CTD kinases. Strategies to analyze samples under both denatured/reduced and semi-native conditions are provided. This method represents a simple, direct, and reproducible means to monitor CTD phosphorylation in recombinant substrates utilizing equipment common to molecular biology labs and readily applicable to downstream analyses including immunoblotting and mass spectrometry.

9.
Proc Natl Acad Sci U S A ; 116(49): 24881-24891, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31754034

ABSTRACT

Dependence on the 26S proteasome is an Achilles' heel for triple-negative breast cancer (TNBC) and multiple myeloma (MM). The therapeutic proteasome inhibitor, bortezomib, successfully targets MM but often leads to drug-resistant disease relapse and fails in breast cancer. Here we show that a 26S proteasome-regulating kinase, DYRK2, is a therapeutic target for both MM and TNBC. Genome editing or small-molecule mediated inhibition of DYRK2 significantly reduces 26S proteasome activity, bypasses bortezomib resistance, and dramatically delays in vivo tumor growth in MM and TNBC thereby promoting survival. We further characterized the ability of LDN192960, a potent and selective DYRK2-inhibitor, to alleviate tumor burden in vivo. The drug docks into the active site of DYRK2 and partially inhibits all 3 core peptidase activities of the proteasome. Our results suggest that targeting 26S proteasome regulators will pave the way for therapeutic strategies in MM and TNBC.


Subject(s)
Bortezomib/pharmacology , Neoplastic Processes , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , TYK2 Kinase/metabolism , Triple Negative Breast Neoplasms/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , Animals , Cell Line, Tumor , Female , Gene Editing , Gene Expression Regulation , Gene Knockout Techniques , HEK293 Cells , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Multiple Myeloma , Phosphorylation , Proteasome Endopeptidase Complex/genetics , Proteasome Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Triple Negative Breast Neoplasms/pathology , Dyrk Kinases
10.
Elife ; 82019 08 06.
Article in English | MEDLINE | ID: mdl-31385803

ABSTRACT

The Positive Transcription Elongation Factor b (P-TEFb) phosphorylates Ser2 residues of the C-terminal domain (CTD) of the largest subunit (RPB1) of RNA polymerase II and is essential for the transition from transcription initiation to elongation in vivo. Surprisingly, P-TEFb exhibits Ser5 phosphorylation activity in vitro. The mechanism garnering Ser2 specificity to P-TEFb remains elusive and hinders understanding of the transition from transcription initiation to elongation. Through in vitro reconstruction of CTD phosphorylation, mass spectrometry analysis, and chromatin immunoprecipitation sequencing (ChIP-seq) analysis, we uncover a mechanism by which Tyr1 phosphorylation directs the kinase activity of P-TEFb and alters its specificity from Ser5 to Ser2. The loss of Tyr1 phosphorylation causes an accumulation of RNA polymerase II in the promoter region as detected by ChIP-seq. We demonstrate the ability of Tyr1 phosphorylation to generate a heterogeneous CTD modification landscape that expands the CTD's coding potential. These findings provide direct experimental evidence for a combinatorial CTD phosphorylation code wherein previously installed modifications direct the identity and abundance of subsequent coding events by influencing the behavior of downstream enzymes.


Subject(s)
Positive Transcriptional Elongation Factor B/metabolism , Protein Processing, Post-Translational , RNA Polymerase II/metabolism , Serine/metabolism , Tyrosine/metabolism , Humans , Phosphorylation , Transcription, Genetic
11.
Neuropharmacology ; 146: 289-299, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30419244

ABSTRACT

Alcohol use disorder (AUD) and major depressive disorder (MDD) are prevalent, debilitating, and highly comorbid disorders. The molecular changes that underlie their comorbidity are beginning to emerge. For example, recent evidence showed that acute ethanol exposure produces rapid antidepressant-like biochemical and behavioral responses. Both ethanol and fast-acting antidepressants block N-methyl-D-aspartate receptor (NMDAR) activity, leading to synaptic changes and long-lasting antidepressant-like behavioral effects. We used RNA sequencing to analyze changes in the synaptic transcriptome after acute treatment with ethanol or the NMDAR antagonist, Ro 25-6981. Ethanol and Ro 25-6981 induced differential, independent changes in gene expression. In contrast with gene-level expression, ethanol and Ro 25-6981 produced overlapping changes in exons, as measured by analysis of differentially expressed exons (DEEs). A prominent overlap in genes with DEEs indicated that changes in exon usage were important for both ethanol and Ro 25-6981 action. Structural modeling provided evidence that ethanol-induced exon expression in the NMDAR1 amino-terminal domain could induce conformational changes and thus alter NMDAR function. These findings suggest that the rapid antidepressant effects of ethanol and NMDAR antagonists reported previously may depend on synaptic exon usage rather than gene expression.


Subject(s)
Alcoholism/genetics , Depressive Disorder, Major/genetics , Exons/drug effects , Exons/genetics , Gene Expression/drug effects , Alternative Splicing/drug effects , Alternative Splicing/genetics , Animals , Antidepressive Agents/pharmacology , Comorbidity , Ethanol/pharmacology , Hippocampus/drug effects , Male , Mice, Inbred C57BL , Models, Animal , Phenols/pharmacology , Piperidines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, Neurotransmitter , Transcriptome
12.
Elife ; 72018 12 06.
Article in English | MEDLINE | ID: mdl-30520731

ABSTRACT

Ca2+ signaling is important for many cellular and physiological processes, including cardiac function. Although sarcoplasmic reticulum (SR) proteins involved in Ca2+ signaling have been shown to be phosphorylated, the biochemical and physiological roles of protein phosphorylation within the lumen of the SR remain essentially uncharacterized. Our laboratory recently identified an atypical protein kinase, Fam20C, which is uniquely localized to the secretory pathway lumen. Here, we show that Fam20C phosphorylates several SR proteins involved in Ca2+ signaling, including calsequestrin2 and Stim1, whose biochemical activities are dramatically regulated by Fam20C mediated phosphorylation. Notably, phosphorylation of Stim1 by Fam20C enhances Stim1 activation and store-operated Ca2+ entry. Physiologically, mice with Fam20c ablated in cardiomyocytes develop heart failure following either aging or induced pressure overload. We extended these observations to show that non-muscle cells lacking Fam20C display altered ER Ca2+ signaling. Overall, we show that Fam20C plays an overarching role in ER/SR Ca2+ homeostasis and cardiac pathophysiology.


Subject(s)
Calcium-Binding Proteins/genetics , Calsequestrin/genetics , Extracellular Matrix Proteins/genetics , Heart Failure/genetics , Stromal Interaction Molecule 1/genetics , Animals , Calcium/chemistry , Calcium/metabolism , Calcium Signaling/genetics , Calcium-Binding Proteins/chemistry , Calsequestrin/chemistry , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/genetics , Extracellular Matrix Proteins/chemistry , Heart Failure/pathology , Homeostasis , Humans , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphorylation , Phosphotransferases/genetics , Sarcoplasmic Reticulum/chemistry , Sarcoplasmic Reticulum/genetics , Secretory Pathway/genetics , Stromal Interaction Molecule 1/chemistry
13.
J Biol Chem ; 293(43): 16851-16861, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30217818

ABSTRACT

The RE1-silencing transcription factor (REST) is the major scaffold protein for assembly of neuronal gene silencing complexes that suppress gene transcription through regulating the surrounding chromatin structure. REST represses neuronal gene expression in stem cells and non-neuronal cells, but it is minimally expressed in neuronal cells to ensure proper neuronal development. Dysregulation of REST function has been implicated in several cancers and neurological diseases. Modulating REST gene silencing is challenging because cellular and developmental differences can affect its activity. We therefore considered the possibility of modulating REST activity through its regulatory proteins. The human small C-terminal domain phosphatase 1 (SCP1) regulates the phosphorylation state of REST at sites that function as REST degradation checkpoints. Using kinetic analysis and direct visualization with X-ray crystallography, we show that SCP1 dephosphorylates two degron phosphosites of REST with a clear preference for phosphoserine 861 (pSer-861). Furthermore, we show that SCP1 stabilizes REST protein levels, which sustains REST's gene silencing function in HEK293 cells. In summary, our findings strongly suggest that REST is a bona fide substrate for SCP1 in vivo and that SCP1 phosphatase activity protects REST against degradation. These observations indicate that targeting REST via its regulatory protein SCP1 can modulate its activity and alter signaling in this essential developmental pathway.


Subject(s)
Neurons/metabolism , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Amino Acid Motifs , Crystallography, X-Ray , Gene Silencing , HEK293 Cells , Humans , Kinetics , Neurons/chemistry , Nuclear Proteins/genetics , Phosphoprotein Phosphatases/genetics , Phosphorylation , Protein Stability , Proteolysis , Repressor Proteins/genetics
14.
Proc Natl Acad Sci U S A ; 115(32): 8155-8160, 2018 08 07.
Article in English | MEDLINE | ID: mdl-29987021

ABSTRACT

Curcumin, the active ingredient in Curcuma longa, has been in medicinal use since ancient times. However, the therapeutic targets and signaling cascades modulated by curcumin have been enigmatic despite extensive research. Here we identify dual-specificity tyrosine-regulated kinase 2 (DYRK2), a positive regulator of the 26S proteasome, as a direct target of curcumin. Curcumin occupies the ATP-binding pocket of DYRK2 in the cocrystal structure, and it potently and specifically inhibits DYRK2 over 139 other kinases tested in vitro. As a result, curcumin diminishes DYRK2-mediated 26S proteasome phosphorylation in cells, leading to reduced proteasome activity and impaired cell proliferation. Interestingly, curcumin synergizes with the therapeutic proteasome inhibitor carfilzomib to induce apoptosis in a variety of proteasome-addicted cancer cells, while this drug combination exhibits modest to no cytotoxicity to noncancerous cells. In a breast cancer xenograft model, curcumin treatment significantly reduces tumor burden in immunocompromised mice, showing a similar antitumor effect as CRISPR/Cas9-mediated DYRK2 depletion. These results reveal an unexpected role of curcumin in DYRK2-proteasome inhibition and provide a proof-of-concept that pharmacological manipulation of proteasome regulators may offer new opportunities for anticancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/pharmacology , Neoplasms/drug therapy , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , CRISPR-Cas Systems , Cell Proliferation/drug effects , Crystallography, X-Ray , Curcumin/therapeutic use , Drug Synergism , Female , Gene Editing/methods , Gene Knockout Techniques/methods , HEK293 Cells , Humans , Inhibitory Concentration 50 , Mice , Neoplasms/pathology , Oligopeptides/pharmacology , Proteasome Inhibitors/pharmacology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Dyrk Kinases
15.
Nat Commun ; 8: 15231, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28497792

ABSTRACT

RNA polymerase II contains a repetitive, intrinsically disordered, C-terminal domain (CTD) composed of heptads of the consensus sequence YSPTSPS. The CTD is heavily phosphorylated and serves as a scaffold, interacting with factors involved in transcription initiation, elongation and termination, RNA processing and chromatin modification. Despite being a nexus of eukaryotic gene regulation, the structure of the CTD and the structural implications of phosphorylation are poorly understood. Here we present a biophysical and biochemical interrogation of the structure of the full length CTD of Drosophila melanogaster, which we conclude is a compact random coil. Surprisingly, we find that the repetitive CTD is structurally heterogeneous. Phosphorylation causes increases in radius, protein accessibility and stiffness, without disrupting local structural heterogeneity. Additionally, we show the human CTD is also structurally heterogeneous and able to substitute for the D. melanogaster CTD in supporting fly development to adulthood. This finding implicates conserved structural organization, not a precise array of heptad motifs, as important to CTD function.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Intrinsically Disordered Proteins/metabolism , RNA Polymerase II/metabolism , Amino Acid Sequence , Animals , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation , Humans , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Models, Molecular , Phosphorylation , Protein Conformation , RNA Polymerase II/chemistry , RNA Polymerase II/genetics , Transcription, Genetic
16.
ACS Chem Biol ; 12(1): 153-162, 2017 01 20.
Article in English | MEDLINE | ID: mdl-28103682

ABSTRACT

Phosphorylation of the C-terminal domain of RNA polymerase II (CTD) plays an essential role in eukaryotic transcription by recruiting transcriptional regulatory factors to the active polymerase. However, the scarcity of basic residues and repetitive nature of the CTD sequence impose a huge challenge for site-specific characterization of phosphorylation, hindering our understanding of this crucial biological process. Herein, we apply LC-UVPD-MS methods to analyze post-translational modification along native sequence CTDs. Application of our method to the Drosophila melanogaster CTD reveals the phosphorylation pattern of this model organism for the first time. The divergent nature of fly CTD allows us to derive rules defining how flanking residues affect phosphorylation choice by CTD kinases. Our data support the use of LC-UVPD-MS to decipher the CTD code and determine rules that program its function.


Subject(s)
Drosophila melanogaster/enzymology , Mass Spectrometry/methods , RNA Polymerase II/metabolism , Amino Acid Sequence , Animals , Drosophila melanogaster/chemistry , Drosophila melanogaster/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Phosphorylation , Protein Domains , Protein Processing, Post-Translational , RNA Polymerase II/chemistry , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/metabolism , Ultraviolet Rays
17.
Biochim Biophys Acta ; 1864(4): 372-87, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26779935

ABSTRACT

The phosphorylation state of the C-terminal domain of RNA polymerase II is required for the temporal and spatial recruitment of various factors that mediate transcription and RNA processing throughout the transcriptional cycle. Therefore, changes in CTD phosphorylation by site-specific kinases/phosphatases are critical for the accurate transmission of information during transcription. Unlike kinases, CTD phosphatases have been traditionally neglected as they are thought to act as passive negative regulators that remove all phosphate marks at the conclusion of transcription. This over-simplified view has been disputed in recent years and new data assert the active and regulatory role phosphatases play in transcription. We now know that CTD phosphatases ensure the proper transition between different stages of transcription, balance the distribution of phosphorylation for accurate termination and re-initiation, and prevent inappropriate expression of certain genes. In this review, we focus on the specific roles of CTD phosphatases in regulating transcription. In particular, we emphasize how specificity and timing of dephosphorylation are achieved for these phosphatases and consider the various regulatory factors that affect these dynamics.


Subject(s)
Phosphoprotein Phosphatases/physiology , RNA Polymerase II/metabolism , Eukaryotic Cells/enzymology , Phosphorylation , Saccharomyces cerevisiae/enzymology , Transcription, Genetic
18.
ACS Chem Biol ; 10(10): 2405-14, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26332362

ABSTRACT

Proline isomerization greatly impacts biological signaling but is subtle and difficult to detect in proteins. We characterize this poorly understood regulatory mechanism for RNA polymerase II carboxyl terminal domain (CTD) phosphorylation state using novel, direct, and quantitative chemical tools. We determine the proline isomeric preference of three CTD phosphatases: Ssu72 as cis-proline specific, Scp1 and Fcp1 as strongly trans-preferred. Due to this inherent characteristic, these phosphatases respond differently to enzymes that catalyze the isomerization of proline, like Ess1/Pin1. We demonstrate that this selective regulation of RNA polymerase II phosphorylation state exists within human cells, consistent with in vitro assays. These results support a model in which, instead of a global enhancement of downstream enzymatic activities, proline isomerases selectively boost the activity of a subset of CTD regulatory factors specific for cis-proline. This leads to diversified phosphorylation states of CTD in vitro and in cells. We provide the chemical tools to investigate proline isomerization and its ability to selectively enhance signaling in transcription and other biological contexts.


Subject(s)
Models, Biological , Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/metabolism , Proline/chemistry , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Animals , Blotting, Western , Crystallography, X-Ray , Drosophila/enzymology , Enzyme Activation/physiology , Humans , Isomerism , Molecular Structure
19.
Molecules ; 19(2): 1481-511, 2014 Jan 27.
Article in English | MEDLINE | ID: mdl-24473209

ABSTRACT

Post-translational modifications of the heptad repeat sequences in the C-terminal domain (CTD) of RNA polymerase II (Pol II) are well recognized for their roles in coordinating transcription with other nuclear processes that impinge upon transcription by the Pol II machinery; and this is primarily achieved through CTD interactions with the various nuclear factors. The identification of novel modifications on new regulatory sites of the CTD suggests that, instead of an independent action for all modifications on CTD, a combinatorial effect is in operation. In this review we focus on two well-characterized modifications of the CTD, namely serine phosphorylation and prolyl isomerization, and discuss the complex interplay between the enzymes modifying their respective regulatory sites. We summarize the current understanding of how the prolyl isomerization state of the CTD dictates the specificity of writers (CTD kinases), erasers (CTD phosphatases) and readers (CTD binding proteins) and how that correlates to transcription status. Subtle changes in prolyl isomerization states cannot be detected at the primary sequence level, we describe the methods that have been utilized to investigate this mode of regulation. Finally, a general model of how prolyl isomerization regulates the phosphorylation state of CTD, and therefore transcription-coupled processes, is proposed.


Subject(s)
Protein Processing, Post-Translational/genetics , RNA Polymerase II/genetics , Repetitive Sequences, Amino Acid/genetics , Transcription, Genetic , Phosphorylation , Phosphotransferases/chemistry , Phosphotransferases/genetics , Protein Binding , Protein Structure, Tertiary , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Saccharomyces cerevisiae/genetics , Serine/chemistry , Serine/genetics
20.
FEBS J ; 280(19): 4739-60, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23937612

ABSTRACT

Protein phosphatases, as the counterpart to protein kinases, are essential for homeostatic balance of cell signaling. Small chemical compounds that modulate the specific activity of phosphatases can be powerful tools to elucidate the biological functions of these enzymes. More importantly, many phosphatases are central players in the development of pathological pathways where inactivation can reverse or delay the onset of human diseases. Therefore, potent inhibitors for such phosphatases can be of great therapeutic benefit. In contrast to the seemingly identical enzymatic mechanism and structural characterization of eukaryotic protein kinases, protein phosphatases evolved from diverse ancestors, resulting in different domain architectures, reaction mechanisms and active site properties. In this review, we discuss for each family of serine/threonine protein phosphatases their involvement in biological processes and corresponding strategies for small chemical intervention. Recent advances in modern drug discovery technologies have markedly facilitated the identification of selective inhibitors for some members of the phosphatase family. Furthermore, the rapid growth in knowledge about structure-activity relationships related to possible new drug targets has aided the discovery of natural product inhibitors for the phosphatase family. This review summarizes the current state of investigation of the small molecules that regulate the function of serine/threonine phosphatases, the challenges presented and also strategies to overcome these obstacles.


Subject(s)
Drug Design , Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/metabolism , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Phosphoprotein Phosphatases/antagonists & inhibitors , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...