Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 423
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 168(6): 1015-1027.e10, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28283058

ABSTRACT

Eukaryotic mRNAs generally possess a 5' end N7 methyl guanosine (m7G) cap that promotes their translation and stability. However, mammalian mRNAs can also carry a 5' end nicotinamide adenine dinucleotide (NAD+) cap that, in contrast to the m7G cap, does not support translation but instead promotes mRNA decay. The mammalian and fungal noncanonical DXO/Rai1 decapping enzymes efficiently remove NAD+ caps, and cocrystal structures of DXO/Rai1 with 3'-NADP+ illuminate the molecular mechanism for how the "deNADding" reaction produces NAD+ and 5' phosphate RNA. Removal of DXO from cells increases NAD+-capped mRNA levels and enables detection of NAD+-capped intronic small nucleolar RNAs (snoRNAs), suggesting NAD+ caps can be added to 5'-processed termini. Our findings establish NAD+ as an alternative mammalian RNA cap and DXO as a deNADding enzyme modulating cellular levels of NAD+-capped RNAs. Collectively, these data reveal that mammalian RNAs can harbor a 5' end modification distinct from the classical m7G cap that promotes rather than inhibits RNA decay.


Subject(s)
RNA Processing, Post-Transcriptional , RNA Stability , Animals , Endoribonucleases/metabolism , HEK293 Cells , Humans , Mice , NAD/metabolism , Nuclear Proteins/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , RNA, Untranslated/metabolism
2.
Cell ; 168(5): 878-889.e29, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28235199

ABSTRACT

Design of small molecules that disrupt protein-protein interactions, including the interaction of RAS proteins and their effectors, may provide chemical probes and therapeutic agents. We describe here the synthesis and testing of potential small-molecule pan-RAS ligands, which were designed to interact with adjacent sites on the surface of oncogenic KRAS. One compound, termed 3144, was found to bind to RAS proteins using microscale thermophoresis, nuclear magnetic resonance spectroscopy, and isothermal titration calorimetry and to exhibit lethality in cells partially dependent on expression of RAS proteins. This compound was metabolically stable in liver microsomes and displayed anti-tumor activity in xenograft mouse cancer models. These findings suggest that pan-RAS inhibition may be an effective therapeutic strategy for some cancers and that structure-based design of small molecules targeting multiple adjacent sites to create multivalent inhibitors may be effective for some proteins.


Subject(s)
Antineoplastic Agents/pharmacology , Molecular Targeted Therapy , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/chemistry , Animals , Antineoplastic Agents/chemistry , Calorimetry , Cell Line , Fibroblasts/metabolism , Heterografts , Humans , Mice , Neoplasm Transplantation , Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Signal Transduction , Small Molecule Libraries
3.
Mol Cell ; 83(3): 416-427, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36634676

ABSTRACT

Integrator is a metazoan-specific protein complex capable of inducing termination at all RNAPII-transcribed loci. Integrator recognizes paused, promoter-proximal RNAPII and drives premature termination using dual enzymatic activities: an endonuclease that cleaves nascent RNA and a protein phosphatase that removes stimulatory phosphorylation associated with RNAPII pause release and productive elongation. Recent breakthroughs in structural biology have revealed the overall architecture of Integrator and provided insights into how multiple Integrator modules are coordinated to elicit termination effectively. Furthermore, functional genomics and biochemical studies have unraveled how Integrator-mediated termination impacts protein-coding and noncoding loci. Here, we review the current knowledge about the assembly and activity of Integrator and describe the role of Integrator in gene regulation, highlighting the importance of this complex for human health.


Subject(s)
Gene Expression Regulation , RNA Polymerase II , Animals , Humans , Phosphoprotein Phosphatases/genetics , Phosphorylation , RNA Polymerase II/metabolism , Transcription, Genetic , Proteins/genetics , Proteins/metabolism
4.
Genes Dev ; 37(21-24): 968-983, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-37977822

ABSTRACT

The spliceosomal gene SF3B1 is frequently mutated in cancer. While it is known that SF3B1 hotspot mutations lead to loss of splicing factor SUGP1 from spliceosomes, the cancer-relevant SF3B1-SUGP1 interaction has not been characterized. To address this issue, we show by structural modeling that two regions flanking the SUGP1 G-patch make numerous contacts with the region of SF3B1 harboring hotspot mutations. Experiments confirmed that all the cancer-associated mutations in these regions, as well as mutations affecting other residues in the SF3B1-SUGP1 interface, not only weaken or disrupt the interaction but also alter splicing similarly to SF3B1 cancer mutations. Finally, structural modeling of a trimeric protein complex reveals that the SF3B1-SUGP1 interaction "loops out" the G-patch for interaction with the helicase DHX15. Our study thus provides an unprecedented molecular view of a protein complex essential for accurate splicing and also reveals that numerous cancer-associated mutations disrupt the critical SF3B1-SUGP1 interaction.


Subject(s)
Neoplasms , Spliceosomes , Humans , RNA, Messenger/metabolism , Spliceosomes/genetics , Spliceosomes/metabolism , RNA Splicing Factors/chemistry , RNA Splicing/genetics , Neoplasms/genetics , Neoplasms/metabolism , Mutation , Phosphoproteins/metabolism
5.
Cell ; 158(6): 1389-1401, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25215494

ABSTRACT

Cyclic di-adenosine monophosphate (c-di-AMP) is a broadly conserved second messenger required for bacterial growth and infection. However, the molecular mechanisms of c-di-AMP signaling are still poorly understood. Using a chemical proteomics screen for c-di-AMP-interacting proteins in the pathogen Listeria monocytogenes, we identified several broadly conserved protein receptors, including the central metabolic enzyme pyruvate carboxylase (LmPC). Biochemical and crystallographic studies of the LmPC-c-di-AMP interaction revealed a previously unrecognized allosteric regulatory site 25 Å from the active site. Mutations in this site disrupted c-di-AMP binding and affected catalytic activity of LmPC as well as PC from pathogenic Enterococcus faecalis. C-di-AMP depletion resulted in altered metabolic activity in L. monocytogenes. Correction of this metabolic imbalance rescued bacterial growth, reduced bacterial lysis, and resulted in enhanced bacterial burdens during infection. These findings greatly expand the c-di-AMP signaling repertoire and reveal a central metabolic regulatory role for a cyclic dinucleotide.


Subject(s)
Dinucleoside Phosphates/metabolism , Listeria monocytogenes/metabolism , Pyruvate Carboxylase/chemistry , Pyruvate Carboxylase/metabolism , Allosteric Regulation , Amino Acid Sequence , Animals , Bacteriolysis , Binding Sites , Crystallography, X-Ray , Host-Pathogen Interactions , Listeria monocytogenes/enzymology , Listeria monocytogenes/growth & development , Listeriosis/microbiology , Mice , Models, Molecular , Molecular Sequence Data
6.
Mol Cell ; 77(4): 800-809.e6, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31810758

ABSTRACT

The mammalian pre-mRNA 3'-end-processing machinery consists of cleavage and polyadenylation specificity factor (CPSF), cleavage stimulation factor (CstF), and other proteins, but the overall architecture of this machinery remains unclear. CPSF contains two functionally distinct modules: a cleavage factor (mCF) and a polyadenylation specificity factor (mPSF). Here, we have produced recombinant human CPSF and CstF and examined these factors by electron microscopy (EM). We find that mPSF is the organizational core of the machinery, while the conformations of mCF and CstF and the position of mCF relative to mPSF are highly variable. We have identified by cryo-EM a segment in CPSF100 that tethers mCF to mPSF, and we have named it the PSF interaction motif (PIM). Mutations in the PIM can abolish CPSF formation, indicating that it is a crucial contact in CPSF. We have also obtained reconstructions of mCF and CstF77 by cryo-EM, assembled around the mPSF core.


Subject(s)
Cleavage And Polyadenylation Specificity Factor/chemistry , Cleavage And Polyadenylation Specificity Factor/genetics , Cleavage Stimulation Factor/chemistry , Cryoelectron Microscopy , Humans , Models, Molecular , Mutation , RNA 3' End Processing , RNA Precursors/metabolism , RNA, Messenger/metabolism
7.
Genes Dev ; 34(23-24): 1753-1761, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33122294

ABSTRACT

Most eukaryotic pre-mRNAs must undergo 3'-end cleavage and polyadenylation prior to their export from the nucleus. A large number of proteins in several complexes participate in this 3'-end processing, including cleavage and polyadenylation specificity factor (CPSF) in mammals. The CPSF30 subunit contains five CCCH zinc fingers (ZFs), with ZF2-ZF3 being required for the recognition of the AAUAAA poly(A) signal. ZF4-ZF5 recruits the hFip1 subunit of CPSF, although the details of this interaction have not been characterized. Here we report the crystal structure of human CPSF30 ZF4-ZF5 in complex with residues 161-200 of hFip1 at 1.9 Å resolution, illuminating the molecular basis for their interaction. Unexpectedly, the structure reveals one hFip1 molecule binding to each ZF4 and ZF5, with a conserved mode of interaction. Our mutagenesis studies confirm that the CPSF30-hFip1 complex has 1:2 stoichiometry in vitro. Mutation of each binding site in CPSF30 still allows one copy of hFip1 to bind, while mutation of both sites abrogates binding. Our fluorescence polarization binding assays show that ZF4 has higher affinity for hFip1, with a Kd of 1.8 nM. We also demonstrate that two copies of the catalytic module of poly(A) polymerase (PAP) are recruited by the CPSF30-hFip1 complex in vitro, and both hFip1 binding sites in CPSF30 can support polyadenylation.


Subject(s)
Cleavage And Polyadenylation Specificity Factor/chemistry , Cleavage And Polyadenylation Specificity Factor/metabolism , Models, Molecular , mRNA Cleavage and Polyadenylation Factors/chemistry , Binding Sites , Cleavage And Polyadenylation Specificity Factor/genetics , Escherichia coli/genetics , Humans , Mutation , Protein Binding , Protein Structure, Quaternary , Zinc Fingers/physiology , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism
8.
Immunity ; 46(3): 433-445, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28329705

ABSTRACT

Bacterial and host cyclic dinucleotides (cdNs) mediate cytosolic immune responses through the STING signaling pathway, although evidence suggests that alternative pathways exist. We used cdN-conjugated beads to biochemically isolate host receptors for bacterial cdNs, and we identified the oxidoreductase RECON. High-affinity cdN binding inhibited RECON enzyme activity by simultaneously blocking the substrate and cosubstrate sites, as revealed by structural analyses. During bacterial infection of macrophages, RECON antagonized STING activation by acting as a molecular sink for cdNs. Bacterial infection of hepatocytes, which do not express STING, revealed that RECON negatively regulates NF-κB activation. Loss of RECON activity, via genetic ablation or inhibition by cdNs, increased NF-κB activation and reduced bacterial survival, suggesting that cdN inhibition of RECON promotes a proinflammatory, antibacterial state that is distinct from the antiviral state associated with STING activation. Thus, RECON functions as a cytosolic sensor for bacterial cdNs, shaping inflammatory gene activation via its effects on STING and NF-κB.


Subject(s)
Bacterial Infections/immunology , Bacterial Proteins/immunology , Estradiol Dehydrogenases/immunology , Inflammation/immunology , NF-kappa B/immunology , Animals , Enzyme Activation/immunology , Macrophages/immunology , Mice , Mice, Knockout , NF-kappa B/metabolism , Real-Time Polymerase Chain Reaction
9.
Proc Natl Acad Sci U S A ; 120(8): e2213272120, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36787367

ABSTRACT

Macropinocytosis is an actin-dependent mode of nonselective endocytosis that mediates the uptake of extracellular fluid-phase cargoes. It is now well recognized that tumor cells exploit macropinocytosis to internalize macromolecules that can be catabolized and used to support cell growth and proliferation under nutrient-limiting conditions. Therefore, the identification of molecular mechanisms that control macropinocytosis is fundamental to the understanding of the metabolic adaptive landscape of tumor cells. Here, we report that the acetyl-CoA-producing enzyme, ATP citrate lyase (ACLY), is a key regulator of macropinocytosis and describes a heretofore-unappreciated association of ACLY with the actin cytoskeleton. The cytoskeletal tethering of ACLY is required for the spatially defined acetylation of heterodimeric actin capping protein, which we identify as an essential mediator of the actin remodeling events that drive membrane ruffling and macropinocytosis. Furthermore, we identify a requirement for mitochondrial-derived citrate, an ACLY substrate, for macropinocytosis, and show that mitochondria traffic to cell periphery regions juxtaposed to plasma membrane ruffles. Collectively, these findings establish a mode of metabolite compartmentalization that supports the spatiotemporal modulation of membrane-cytoskeletal interactions required for macropinocytosis by coupling regional acetyl-CoA availability with dynamic protein acetylation.


Subject(s)
ATP Citrate (pro-S)-Lyase , Actins , ATP Citrate (pro-S)-Lyase/genetics , ATP Citrate (pro-S)-Lyase/metabolism , Acetyl Coenzyme A/metabolism , Actins/metabolism , Protein Processing, Post-Translational , Cell Proliferation
10.
RNA ; 29(11): 1673-1690, 2023 11.
Article in English | MEDLINE | ID: mdl-37562960

ABSTRACT

U7 snRNP is a multisubunit endonuclease required for 3' end processing of metazoan replication-dependent histone pre-mRNAs. In contrast to the spliceosomal snRNPs, U7 snRNP lacks the Sm subunits D1 and D2 and instead contains two related proteins, Lsm10 and Lsm11. The remaining five subunits of the U7 heptameric Sm ring, SmE, F, G, B, and D3, are shared with the spliceosomal snRNPs. The pathway that assembles the unique ring of U7 snRNP is unknown. Here, we show that a heterodimer of Lsm10 and Lsm11 tightly interacts with the methylosome, a complex of the arginine methyltransferase PRMT5, MEP50, and pICln known to methylate arginines in the carboxy-terminal regions of the Sm proteins B, D1, and D3 during the spliceosomal Sm ring assembly. Both biochemical and cryo-EM structural studies demonstrate that the interaction is mediated by PRMT5, which binds and methylates two arginine residues in the amino-terminal region of Lsm11. Surprisingly, PRMT5 also methylates an amino-terminal arginine in SmE, a subunit that does not undergo this type of modification during the biogenesis of the spliceosomal snRNPs. An intriguing possibility is that the unique methylation pattern of Lsm11 and SmE plays a vital role in the assembly of the U7 snRNP.


Subject(s)
Ribonucleoprotein, U7 Small Nuclear , Ribonucleoproteins, Small Nuclear , Animals , Ribonucleoprotein, U7 Small Nuclear/chemistry , Methylation , Ribonucleoproteins, Small Nuclear/metabolism , Histones/metabolism , Arginine/chemistry
11.
Nature ; 568(7753): 566-570, 2019 04.
Article in English | MEDLINE | ID: mdl-30944472

ABSTRACT

ATP-citrate lyase (ACLY) is a central metabolic enzyme and catalyses the ATP-dependent conversion of citrate and coenzyme A (CoA) to oxaloacetate and acetyl-CoA1-5. The acetyl-CoA product is crucial for the metabolism of fatty acids6,7, the biosynthesis of cholesterol8, and the acetylation and prenylation of proteins9,10. There has been considerable interest in ACLY as a target for anti-cancer drugs, because many cancer cells depend on its activity for proliferation2,5,11. ACLY is also a target against dyslipidaemia and hepatic steatosis, with a compound currently in phase 3 clinical trials4,5. Many inhibitors of ACLY have been reported, but most of them have weak activity5. Here we report the development of a series of low nanomolar, small-molecule inhibitors of human ACLY. We have also determined the structure of the full-length human ACLY homo-tetramer in complex with one of these inhibitors (NDI-091143) by cryo-electron microscopy, which reveals an unexpected mechanism of inhibition. The compound is located in an allosteric, mostly hydrophobic cavity next to the citrate-binding site, and requires extensive conformational changes in the enzyme that indirectly disrupt citrate binding. The observed binding mode is supported by and explains the structure-activity relationships of these compounds. This allosteric site greatly enhances the 'druggability' of ACLY and represents an attractive target for the development of new ACLY inhibitors.


Subject(s)
ATP Citrate (pro-S)-Lyase/antagonists & inhibitors , ATP Citrate (pro-S)-Lyase/ultrastructure , Benzyl Compounds/pharmacology , Cryoelectron Microscopy , Enzyme Inhibitors/pharmacology , ATP Citrate (pro-S)-Lyase/chemistry , ATP Citrate (pro-S)-Lyase/metabolism , Adenosine Diphosphate/metabolism , Allosteric Regulation/drug effects , Benzyl Compounds/chemistry , Binding Sites/drug effects , Citric Acid/metabolism , Enzyme Inhibitors/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Protein Domains , Protein Multimerization , Structure-Activity Relationship
12.
Proc Natl Acad Sci U S A ; 119(49): e2216712119, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36459648

ABSTRACT

SF3B1 is the most frequently mutated spliceosomal gene in cancer. Several hotspot mutations are known to disrupt the interaction of SF3B1 with another splicing factor, SUGP1, resulting in the RNA missplicing that characterizes mutant SF3B1 cancers. Properties of SUGP1, especially the presence of a G-patch motif, a structure known to function by activating DEAH-box RNA helicases, suggest the requirement of such an enzyme in SUGP1 function in splicing. However, the identity of this putative helicase has remained an important unanswered question. Here, using a variety of protein-protein interaction assays, we identify DHX15 as the critical helicase. We further show that depletion of DHX15 or expression of any of several DHX15 mutants, including one implicated in acute myeloid leukemia, partially recapitulates the splicing defects of mutant SF3B1. Moreover, a DHX15-SUGP1 G-patch fusion protein is able to incorporate into the spliceosome to rescue the splicing defects of mutant SF3B1. We also present the crystal structure of the human DHX15-SUGP1 G-patch complex, which reveals the molecular basis of their direct interaction. Our data thus demonstrate that DHX15 is the RNA helicase that functions with SUGP1 and additionally provide important insight into how mutant SF3B1 disrupts splicing in cancer.


Subject(s)
Neoplasms , RNA Helicases , RNA Splicing Factors , RNA Splicing , Humans , DNA Helicases , Genes, Regulator , Phosphoproteins , RNA Helicases/genetics , RNA Splicing/genetics , RNA Splicing Factors/genetics , Spliceosomes/genetics
13.
J Biol Chem ; 299(4): 103047, 2023 04.
Article in English | MEDLINE | ID: mdl-36822327

ABSTRACT

Human cleavage and polyadenylation specificity factor (CPSF)73 (also known as CPSF3) is the endoribonuclease that catalyzes the cleavage reaction for the 3'-end processing of pre-mRNAs. The active site of CPSF73 is located at the interface between a metallo-ß-lactamase domain and a ß-CASP domain. Two metal ions are coordinated by conserved residues, five His and two Asp, in the active site, and they are critical for the nuclease reaction. The metal ions have long been thought to be zinc ions, but their exact identity has not been examined. Here we present evidence from inductively coupled plasma mass spectrometry and X-ray diffraction analyses that a mixture of metal ions, including Fe, Zn, and Mn, is present in the active site of CPSF73. The abundance of the various metal ions is different in samples prepared from different expression hosts. Zinc is present at less than 20% abundance in a sample expressed in insect cells, but the sample is active in cleaving a pre-mRNA substrate in a reconstituted canonical 3'-end processing machinery. Zinc is present at 75% abundance in a sample expressed in human cells, which has comparable endonuclease activity. We also observe a mixture of metal ions in the active site of the CPSF73 homolog INTS11, the endonuclease for Integrator. Taken together, our results provide further insights into the role of metal ions in the activity of CPSF73 and INTS11 for RNA 3'-end processing.


Subject(s)
Cleavage And Polyadenylation Specificity Factor , Endonucleases , Humans , Catalytic Domain , Cleavage And Polyadenylation Specificity Factor/chemistry , Cleavage And Polyadenylation Specificity Factor/metabolism , Endonucleases/chemistry , Endonucleases/metabolism , RNA Processing, Post-Transcriptional , Zinc/metabolism
14.
RNA ; 28(11): 1534-1541, 2022 11.
Article in English | MEDLINE | ID: mdl-36130077

ABSTRACT

The polyadenylation signal (PAS) is a key sequence element for 3'-end cleavage and polyadenylation of messenger RNA precursors (pre-mRNAs). This hexanucleotide motif is recognized by the mammalian polyadenylation specificity factor (mPSF), consisting of CPSF160, WDR33, CPSF30, and Fip1 subunits. Recent studies have revealed how the AAUAAA PAS, the most frequently observed PAS, is recognized by mPSF. We report here the structure of human mPSF in complex with the AUUAAA PAS, the second most frequently identified PAS. Conformational differences are observed for the A1 and U2 nucleotides in AUUAAA compared to the A1 and A2 nucleotides in AAUAAA, while the binding modes of the remaining 4 nt are essentially identical. The 5' phosphate of U2 moves by 2.6 Å and the U2 base is placed near the six-membered ring of A2 in AAUAAA, where it makes two hydrogen bonds with zinc finger 2 (ZF2) of CPSF30, which undergoes conformational changes as well. We also attempted to determine the binding modes of two rare PAS hexamers, AAGAAA and GAUAAA, but did not observe the RNA in the cryo-electron microscopy density. The residues in CPSF30 (ZF2 and ZF3) and WDR33 that recognize PAS are disordered in these two structures.


Subject(s)
Polyadenylation , mRNA Cleavage and Polyadenylation Factors , Animals , Humans , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism , Cleavage And Polyadenylation Specificity Factor/metabolism , Cryoelectron Microscopy , RNA, Messenger/metabolism , Protein Binding , RNA Precursors/metabolism , Mammals/genetics , Nucleotides/metabolism , Poly A/metabolism
15.
Macromol Rapid Commun ; : e2400093, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38639102

ABSTRACT

The formation of ABC triblock terpolymers through solution casting is still challenging. In this study, core-shell double gyroid network structures are fabricated via solution casting using poly(2,2,2-trifluoroethyl methacrylate) (PTFEMA) (F)-b-[poly(4-vinylpyridine) (P4VP) (P)]-b-[polystyrene (PS) (S)] (FPS) triblock terpolymers in N,N-dimethylformamide (DMF). Upon heat treatment, the polymer tends to form a sphere-in-lamellar structure at the F/S interface. Given the solubility properties of each component in DMF, it is anticipated that the effective volume fraction of F relative to P would increase in concentrated solutions and the effective volume fraction of S would decrease. The microphase-separated structure derived from the DMF solution consistently results in the formation of a network structure composed of a core-shell double gyroid, with F as the matrix, P as the shell, and S as the core, and their periodic lengths gradually increase to 110.8, 131.8, and 162.7 nm as increase molecular weights of PS blocks to 13.8, 20.7, and 28.8 kg mol-1. Based on the solubility properties of the polymer components highlighted in this study, the solvent selection strategy is broadly applicable to ABC triblock terpolymers featuring various polymer components, offering a more efficient avenue for fabricating core-shell double gyroid structures.

16.
J Biopharm Stat ; : 1-17, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38506674

ABSTRACT

Multi-regional clinical trial (MRCT) has become an increasing trend for its supporting simultaneous global drug development. After MRCT, consistency assessment needs to be conducted to evaluate regional efficacy. The weighted Z-test approach is a common consistency assessment approach in which the weighting parameter W does not have a good practical significance; the discounting factor approach improved from the weighted Z-test approach by converting the estimation of W in original weighted Z-test approach to the estimation of discounting factor D. However, the discounting factor approach is an approach of frequency statistics, in which D was fixed as a certain value; the variation of D was not considered, which may lead to un-reasonable results. In this paper, we proposed a Bayesian approach based on D to evaluate the treatment effect for the target region in MRCT, in which the variation of D was considered. Specifically, we first took D random instead of fixed as a certain value and specified a beta distribution for it. According to the results of simulation, we further adjusted the Bayesian approach. The application of the proposed approach was illustrated by Markov Chain Monte Carlo simulation.

17.
Nucleic Acids Res ; 50(15): 8807-8817, 2022 08 26.
Article in English | MEDLINE | ID: mdl-35904778

ABSTRACT

Identification of metabolite caps including FAD on the 5' end of RNA has uncovered a previously unforeseen intersection between cellular metabolism and gene expression. To understand the function of FAD caps in cellular physiology, we characterised the proteins interacting with FAD caps in budding yeast. Here we demonstrate that highly conserved 5'-3' exoribonucleases, Xrn1 and Rat1, physically interact with the RNA 5' FAD cap and both possess FAD cap decapping (deFADding) activity and subsequently degrade the resulting RNA. Xrn1 deFADding activity was also evident in human cells indicating its evolutionary conservation. Furthermore, we report that the recently identified bacterial 5'-3' exoribonuclease RNase AM also possesses deFADding activity that can degrade FAD-capped RNAs in vitro and in Escherichia coli cells. To gain a molecular understanding of the deFADding reaction, an RNase AM crystal structure with three manganese ions coordinated by a sulfate molecule and the active site amino acids was generated that provided details underlying hydrolysis of the FAD cap. Our findings reveal a general propensity for 5'-3' exoribonucleases to hydrolyse and degrade RNAs with 5' end noncanonical caps in addition to their well characterized 5' monophosphate RNA substrates indicating an intrinsic property of 5'-3' exoribonucleases.


Subject(s)
Exoribonucleases , Saccharomyces cerevisiae Proteins , Exoribonucleases/metabolism , Flavin-Adenine Dinucleotide/metabolism , Humans , RNA Caps/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism
18.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33649224

ABSTRACT

A number of plant-associated proteobacteria have LuxR family transcription factors that we refer to as PipR subfamily members. PipR proteins play roles in interactions between bacteria and their plant hosts, and some are important for bacterial virulence of plants. We identified an ethanolamine derivative, N-(2-hydroxyethyl)-2-(2-hydroxyethylamino) acetamide (HEHEAA), as a potent effector of PipR-mediated gene regulation in the plant endophyte Pseudomonas GM79. HEHEAA-dependent PipR activity requires an ATP-binding cassette-type active transport system, and the periplasmic substrate-binding protein (SBP) of that system binds HEHEAA. To begin to understand the molecular basis of PipR system responses to plant factors we crystallized a HEHEAA-responsive SBP in the free- and HEHEAA-bound forms. The SBP, which is similar to peptide-binding SBPs, was in a closed conformation. A narrow cavity at the interface of its two lobes is wide enough to bind HEHEAA, but it cannot accommodate peptides with side chains. The polar atoms of HEHEAA are recognized by hydrogen-bonding interactions, and additional SBP residues contribute to the binding site. This binding mode was confirmed by a structure-based mutational analysis. We also show that a closely related SBP from the plant pathogen Pseudomonas syringae pv tomato DC3000 does not recognize HEHEAA. However, a single amino acid substitution in the presumed effector-binding pocket of the P. syringae SBP converted it to a weak HEHEAA-binding protein. The P. syringae PipR depends on a plant effector for activity, and our findings imply that different PipR-associated SBPs bind different effectors.


Subject(s)
Acetamides/chemistry , Bacterial Proteins/chemistry , Pseudomonas syringae/chemistry , Acetamides/metabolism , Bacterial Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Pseudomonas syringae/metabolism
19.
20.
Genes Dev ; 30(1): 117-31, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26728557

ABSTRACT

The transcription cycle of RNA polymerase II (Pol II) is regulated at discrete transition points by cyclin-dependent kinases (CDKs). Positive transcription elongation factor b (P-TEFb), a complex of Cdk9 and cyclin T1, promotes release of paused Pol II into elongation, but the precise mechanisms and targets of Cdk9 action remain largely unknown. Here, by a chemical genetic strategy, we identified ∼ 100 putative substrates of human P-TEFb, which were enriched for proteins implicated in transcription and RNA catabolism. Among the RNA processing factors phosphorylated by Cdk9 was the 5'-to-3' "torpedo" exoribonuclease Xrn2, required in transcription termination by Pol II, which we validated as a bona fide P-TEFb substrate in vivo and in vitro. Phosphorylation by Cdk9 or phosphomimetic substitution of its target residue, Thr439, enhanced enzymatic activity of Xrn2 on synthetic substrates in vitro. Conversely, inhibition or depletion of Cdk9 or mutation of Xrn2-Thr439 to a nonphosphorylatable Ala residue caused phenotypes consistent with inefficient termination in human cells: impaired Xrn2 chromatin localization and increased readthrough transcription of endogenous genes. Therefore, in addition to its role in elongation, P-TEFb regulates termination by promoting chromatin recruitment and activation of a cotranscriptional RNA processing enzyme, Xrn2.


Subject(s)
Cyclin-Dependent Kinase 9/metabolism , Exoribonucleases/genetics , Exoribonucleases/metabolism , Gene Expression Regulation/genetics , Positive Transcriptional Elongation Factor B/metabolism , Chromatin/metabolism , Enzyme Activation/genetics , Genetic Testing , HCT116 Cells , Humans , Phosphorylation , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL