Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
2.
Acc Chem Res ; 56(7): 763-775, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36917683

ABSTRACT

RNA molecules convey biological information both in their linear sequence and in their base-paired secondary and tertiary structures. Chemical probing experiments, which involve treating an RNA with a reagent that modifies conformationally dynamic nucleotides, have broadly enabled examination of short- and long-range RNA structure in diverse contexts, including in living cells. For decades, chemical probing experiments have been interpreted in a per-nucleotide way, such that the reactivity measured at each nucleotide reports the average structure at a position over all RNA molecules within a sample. However, there are numerous important cases where per-nucleotide chemical probing falls short, including for RNAs that are bound by proteins, RNAs that form complex higher order structures, and RNAs that sample multiple conformations.Recent experimental and computational innovations have started a revolution in RNA structure analysis by transforming chemical probing into a massively parallel, single-molecule experiment. Enabled by a specialized reverse transcription strategy called mutational profiling (MaP), multiple chemical modification events can be measured within individual RNA molecules. Nucleotides that communicate structurally through direct base pairing or large-scale folding-unfolding transitions will react with chemical probes in a correlated manner, thereby revealing structural complexity hidden to conventional approaches. These single-molecule correlated chemical probing (smCCP) experiments can be interpreted to directly identify nucleotides that base pair (the PAIR-MaP strategy) and to reveal long-range, through-space structural communication (RING-MaP). Correlated probing can also define the thermodynamic populations of complex RNA ensembles (DANCE-MaP). Complex RNA-protein networks can be interrogated by cross-linking proteins to RNA and measuring correlations between cross-linked positions (RNP-MaP).smCCP thus visualizes RNA secondary and higher-order structure with unprecedented accuracy, defining novel structures, RNA-protein interaction networks, time-resolved dynamics, and allosteric structural switches. These strategies are not mutually exclusive; in favorable cases, multiple levels of RNA structure ─ base pairing, through-space structural communication, and equilibrium ensembles ─ can be resolved concurrently. The physical experimentation required for smCCP is profoundly simple, and experiments are readily performed in cells on RNAs of any size, including large noncoding RNAs and mRNAs. Single-molecule correlated chemical probing is paving the way for a new generation of biophysical studies on RNA in living systems.


Subject(s)
Nucleotides , RNA , Nucleic Acid Conformation , RNA/chemistry , Base Pairing , RNA, Messenger , Proteins/genetics
3.
RNA ; 29(5): 691-704, 2023 05.
Article in English | MEDLINE | ID: mdl-36792358

ABSTRACT

Although not canonically polyadenylated, the long noncoding RNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) is stabilized by a highly conserved 76-nt triple helix structure on its 3' end. The entire MALAT1 transcript is over 8000 nt long in humans. The strongest structural conservation signal in MALAT1 (as measured by covariation of base pairs) is in the triple helix structure. Primary sequence analysis of covariation alone does not reveal the degree of structural conservation of the entire full-length transcript, however. Furthermore, RNA structure is often context dependent; RNA binding proteins that are differentially expressed in different cell types may alter structure. We investigate here the in-cell and cell-free structures of the full-length human and green monkey (Chlorocebus sabaeus) MALAT1 transcripts in multiple tissue-derived cell lines using SHAPE chemical probing. Our data reveal levels of uniform structural conservation in different cell lines, in cells and cell-free, and even between species, despite significant differences in primary sequence. The uniformity of the structural conservation across the entire transcript suggests that, despite seeing covariation signals only in the triple helix junction of the lncRNA, the rest of the transcript's structure is remarkably conserved, at least in primates and across multiple cell types and conditions.


Subject(s)
RNA, Long Noncoding , Animals , Humans , Chlorocebus aethiops , RNA, Long Noncoding/metabolism , Base Pairing , Cell Line , RNA Stability , Cell Proliferation , Cell Line, Tumor
4.
Nucleic Acids Res ; 50(17): 9689-9704, 2022 09 23.
Article in English | MEDLINE | ID: mdl-36107773

ABSTRACT

SERPINA1 mRNAs encode the protease inhibitor α-1-antitrypsin and are regulated through post-transcriptional mechanisms. α-1-antitrypsin deficiency leads to chronic obstructive pulmonary disease (COPD) and liver cirrhosis, and specific variants in the 5'-untranslated region (5'-UTR) are associated with COPD. The NM_000295.4 transcript is well expressed and translated in lung and blood and features an extended 5'-UTR that does not contain a competing upstream open reading frame (uORF). We show that the 5'-UTR of NM_000295.4 folds into a well-defined multi-helix structural domain. We systematically destabilized mRNA structure across the NM_000295.4 5'-UTR, and measured changes in (SHAPE quantified) RNA structure and cap-dependent translation relative to a native-sequence reporter. Surprisingly, despite destabilizing local RNA structure, most mutations either had no effect on or decreased translation. Most structure-destabilizing mutations retained native, global 5'-UTR structure. However, those mutations that disrupted the helix that anchors the 5'-UTR domain yielded three groups of non-native structures. Two of these non-native structure groups refolded to create a stable helix near the translation initiation site that decreases translation. Thus, in contrast to the conventional model that RNA structure in 5'-UTRs primarily inhibits translation, complex folding of the NM_000295.4 5'-UTR creates a translation-optimized message by promoting accessibility at the translation initiation site.


Subject(s)
Protein Biosynthesis , Pulmonary Disease, Chronic Obstructive , alpha 1-Antitrypsin/genetics , 5' Untranslated Regions , Humans , Protease Inhibitors , Pulmonary Disease, Chronic Obstructive/genetics , RNA, Messenger/metabolism
5.
Mol Cell ; 82(9): 1708-1723.e10, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35320755

ABSTRACT

7SK is a conserved noncoding RNA that regulates transcription by sequestering the transcription factor P-TEFb. 7SK function entails complex changes in RNA structure, but characterizing RNA dynamics in cells remains an unsolved challenge. We developed a single-molecule chemical probing strategy, DANCE-MaP (deconvolution and annotation of ribonucleic conformational ensembles), that defines per-nucleotide reactivity, direct base pairing interactions, tertiary interactions, and thermodynamic populations for each state in RNA structural ensembles from a single experiment. DANCE-MaP reveals that 7SK RNA encodes a large-scale structural switch that couples dissolution of the P-TEFb binding site to structural remodeling at distal release factor binding sites. The 7SK structural equilibrium shifts in response to cell growth and stress and can be targeted to modulate expression of P-TEFbresponsive genes. Our study reveals that RNA structural dynamics underlie 7SK function as an integrator of diverse cellular signals to control transcription and establishes the power of DANCE-MaP to define RNA dynamics in cells.


Subject(s)
Positive Transcriptional Elongation Factor B , RNA-Binding Proteins , Binding Sites/genetics , HeLa Cells , Humans , Positive Transcriptional Elongation Factor B/genetics , RNA, Small Nuclear/genetics , RNA, Untranslated , RNA-Binding Proteins/genetics
6.
Cell Rep ; 38(7): 110361, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35172143

ABSTRACT

Many lncRNAs have been discovered using transcriptomic data; however, it is unclear what fraction of lncRNAs is functional and what structural properties affect their phenotype. MUNC lncRNA (also known as DRReRNA) acts as an enhancer RNA for the Myod1 gene in cis and stimulates the expression of other promyogenic genes in trans by recruiting the cohesin complex. Here, experimental probing of the RNA structure revealed that MUNC contains multiple structural domains not detected by prediction algorithms in the absence of experimental information. We show that these specific and structurally distinct domains are required for induction of promyogenic genes, for binding genomic sites and gene expression regulation, and for binding the cohesin complex. Myod1 induction and cohesin interaction comprise only a subset of MUNC phenotype. Our study reveals unexpectedly complex, structure-driven functions for the MUNC lncRNA and emphasizes the importance of experimentally determined structures for understanding structure-function relationships in lncRNAs.


Subject(s)
Muscle Development/genetics , RNA, Long Noncoding/metabolism , Transcription, Genetic , Animals , Base Sequence , Cell Differentiation/genetics , Cell Line , Female , Genome , Mice , Muscle Fibers, Skeletal/metabolism , Nucleic Acid Conformation , Phenotype , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Long Noncoding/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Deletion
7.
Nat Biotechnol ; 39(3): 347-356, 2021 03.
Article in English | MEDLINE | ID: mdl-33077962

ABSTRACT

RNA-protein interaction networks govern many biological processes but are difficult to examine comprehensively. We devised ribonucleoprotein networks analyzed by mutational profiling (RNP-MaP), a live-cell chemical probing strategy that maps cooperative interactions among multiple proteins bound to single RNA molecules at nucleotide resolution. RNP-MaP uses a hetero-bifunctional crosslinker to freeze interacting proteins in place on RNA and then maps multiple bound proteins on single RNA strands by read-through reverse transcription and DNA sequencing. RNP-MaP revealed that RNase P and RMRP, two sequence-divergent but structurally related non-coding RNAs, share RNP networks and that network hubs define functional sites in these RNAs. RNP-MaP also identified protein interaction networks conserved between mouse and human XIST long non-coding RNAs and defined protein communities whose binding sites colocalize and form networks in functional regions of XIST. RNP-MaP enables discovery and efficient validation of functional protein interaction networks on long RNAs in living cells.


Subject(s)
RNA-Binding Proteins/metabolism , RNA/metabolism , Ribonucleoproteins/metabolism , Animals , Humans , Protein Interaction Maps , RNA, Long Noncoding/metabolism , Reproducibility of Results , Ribonuclease P/metabolism
8.
Mol Cell ; 80(6): 1078-1091.e6, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33290746

ABSTRACT

We report that the SARS-CoV-2 nucleocapsid protein (N-protein) undergoes liquid-liquid phase separation (LLPS) with viral RNA. N-protein condenses with specific RNA genomic elements under physiological buffer conditions and condensation is enhanced at human body temperatures (33°C and 37°C) and reduced at room temperature (22°C). RNA sequence and structure in specific genomic regions regulate N-protein condensation while other genomic regions promote condensate dissolution, potentially preventing aggregation of the large genome. At low concentrations, N-protein preferentially crosslinks to specific regions characterized by single-stranded RNA flanked by structured elements and these features specify the location, number, and strength of N-protein binding sites (valency). Liquid-like N-protein condensates form in mammalian cells in a concentration-dependent manner and can be altered by small molecules. Condensation of N-protein is RNA sequence and structure specific, sensitive to human body temperature, and manipulatable with small molecules, and therefore presents a screenable process for identifying antiviral compounds effective against SARS-CoV-2.


Subject(s)
COVID-19/metabolism , Coronavirus Nucleocapsid Proteins/metabolism , Genome, Viral , Nucleocapsid/metabolism , RNA, Viral/metabolism , SARS-CoV-2/metabolism , Animals , Antiviral Agents/pharmacology , COVID-19/genetics , Chlorocebus aethiops , Coronavirus Nucleocapsid Proteins/genetics , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Nucleocapsid/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , SARS-CoV-2/genetics , Vero Cells , COVID-19 Drug Treatment
9.
Cell Rep ; 30(2): 541-554.e5, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31940495

ABSTRACT

Long non-coding RNAs (lncRNAs) are critical regulators of numerous physiological processes and diseases, especially cancers. However, development of lncRNA-based therapies is limited because the mechanisms of many lncRNAs are obscure, and interactions with functional partners, including proteins, remain uncharacterized. The lncRNA SLNCR1 binds to and regulates the androgen receptor (AR) to mediate melanoma invasion and proliferation in an androgen-independent manner. Here, we use biochemical analyses coupled with selective 2'-hydroxyl acylation analyzed by primer extension (SHAPE) RNA structure probing to show that the N-terminal domain of AR binds a pyrimidine-rich motif in an unstructured region of SLNCR1. This motif is predictive of AR binding, as we identify an AR-binding motif in lncRNA HOXA11-AS-203. Oligonucleotides that bind either the AR N-terminal domain or the AR RNA motif block the SLNCR1-AR interaction and reduce SLNCR1-mediated melanoma invasion. Delivery of oligos that block SLNCR1-AR interaction thus represent a plausible therapeutic strategy.


Subject(s)
Melanoma/metabolism , RNA, Long Noncoding/metabolism , Receptors, Androgen/metabolism , Base Sequence , Cell Line, Tumor , Cell Proliferation/physiology , Female , HEK293 Cells , Humans , Male , Melanoma/genetics , Melanoma/pathology , Neoplasm Invasiveness , Protein Domains , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptors, Androgen/genetics
10.
Biochemistry ; 58(31): 3377-3385, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31305988

ABSTRACT

Chemical probing experiments, coupled with empirically determined free energy change relationships, can enable accurate modeling of the secondary structures of diverse and complex RNAs. A current frontier lies in modeling large and structurally heterogeneous transcripts, including complex eukaryotic RNAs. To validate and improve on experimentally driven approaches for modeling large transcripts, we obtained high-quality SHAPE data for the protein-free human 18S and 28S ribosomal RNAs (rRNAs). To our surprise, SHAPE-directed structure models for the human rRNAs poorly matched accepted structures. Analysis of predicted rRNA structures based on low-SHAPE and low-entropy (lowSS) metrics revealed that, whereas ∼75% of Escherichia coli rRNA sequences form well-determined lowSS secondary structure, only ∼40% of the human rRNAs do. Critically, regions of the human rRNAs that specifically fold into well-determined lowSS structures were modeled to high accuracy using SHAPE data. This work reveals that eukaryotic rRNAs are more unfolded than are those of prokaryotic rRNAs and indeed are largely unfolded overall, likely reflecting increased protein dependence for eukaryotic ribosome structure. In addition, those regions and substructures that are well-determined can be identified de novo and successfully modeled by SHAPE-directed folding.


Subject(s)
Nucleic Acid Conformation , RNA, Ribosomal, 18S/chemistry , RNA, Ribosomal, 28S/chemistry , Acylation , Base Sequence , Escherichia coli/genetics , HEK293 Cells , Humans , RNA, Ribosomal, 18S/genetics , RNA, Ribosomal, 28S/genetics , Solutions
11.
Biochemistry ; 58(23): 2655-2664, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31117385

ABSTRACT

Chemical probing is an important tool for characterizing the complex folded structures of RNA molecules, many of which play key cellular roles. Electrophilic SHAPE reagents create adducts at the 2'-hydroxyl position on the RNA backbone of flexible ribonucleotides with relatively little dependence on nucleotide identity. Strategies for adduct detection such as mutational profiling (MaP) allow accurate, automated calculation of relative adduct frequencies for each nucleotide in a given RNA or group of RNAs. A number of alternative reagents and adduct detection strategies have been proposed, especially for use in living cells. Here we evaluate five SHAPE reagents: three previously well-validated reagents 1M7 (1-methyl-7-nitroisatoic anhydride), 1M6 (1-methyl-6-nitroisatoic anhydride), and NMIA ( N-methylisatoic anhydride), one more recently proposed NAI (2-methylnicotinic acid imidazolide), and one novel reagent 5NIA (5-nitroisatoic anhydride). We clarify the importance of carefully designed software in reading out SHAPE experiments using massively parallel sequencing approaches. We examine SHAPE modification in living cells in diverse cell lines, compare MaP and reverse transcription-truncation as SHAPE adduct detection strategies, make recommendations for SHAPE reagent choice, and outline areas for future development.


Subject(s)
Indicators and Reagents/chemistry , Molecular Probes/chemistry , RNA, Bacterial/chemistry , Anhydrides/chemistry , Animals , Escherichia coli/chemistry , High-Throughput Nucleotide Sequencing/methods , Humans , Jurkat Cells , Mice , Oxazines/chemistry , Sequence Analysis, RNA/methods , ortho-Aminobenzoates/chemistry
12.
Science ; 360(6391): 922-927, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29650703

ABSTRACT

RNA promotes liquid-liquid phase separation (LLPS) to build membraneless compartments in cells. How distinct molecular compositions are established and maintained in these liquid compartments is unknown. Here, we report that secondary structure allows messenger RNAs (mRNAs) to self-associate and determines whether an mRNA is recruited to or excluded from liquid compartments. The polyQ-protein Whi3 induces conformational changes in RNA structure and generates distinct molecular fluctuations depending on the RNA sequence. These data support a model in which structure-based, RNA-RNA interactions promote assembly of distinct droplets and protein-driven, conformational dynamics of the RNA maintain this identity. Thus, the shape of RNA can promote the formation and coexistence of the diverse array of RNA-rich liquid compartments found in a single cell.


Subject(s)
Peptides/chemistry , Phase Transition , RNA, Messenger/chemistry , RNA-Binding Proteins/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Base Sequence , Cyclins/chemistry , Nucleic Acid Conformation
13.
RNA Biol ; 14(11): 1445-1456, 2017 11 02.
Article in English | MEDLINE | ID: mdl-28318367

ABSTRACT

Eukaryotes possess a vast array of RNA-binding proteins (RBPs) that affect mRNAs in diverse ways to control protein expression. Combinatorial regulation of mRNAs by RBPs is emerging as the rule. No example illustrates this as vividly as the partnership of 3 Drosophila RBPs, Pumilio, Nanos and Brain Tumor, which have overlapping functions in development, stem cell maintenance and differentiation, fertility and neurologic processes. Here we synthesize 30 y of research with new insights into their molecular functions and mechanisms of action. First, we provide an overview of the key properties of each RBP. Next, we present a detailed analysis of their collaborative regulatory mechanism using a classic example of the developmental morphogen, hunchback, which is spatially and temporally regulated by the trio during embryogenesis. New biochemical, structural and functional analyses provide insights into RNA recognition, cooperativity, and regulatory mechanisms. We integrate these data into a model of combinatorial RNA binding and regulation of translation and mRNA decay. We then use this information, transcriptome wide analyses and bioinformatics predictions to assess the global impact of Pumilio, Nanos and Brain Tumor on gene regulation. Together, the results support pervasive, dynamic post-transcriptional control.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Transcription Factors/genetics , Animals , Binding Sites , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism
14.
Methods ; 118-119: 171-181, 2017 04 15.
Article in English | MEDLINE | ID: mdl-27729296

ABSTRACT

RNA-binding proteins (RBPs) collaborate to control virtually every aspect of RNA function. Tremendous progress has been made in the area of global assessment of RBP specificity using next-generation sequencing approaches both in vivo and in vitro. Understanding how protein-protein interactions enable precise combinatorial regulation of RNA remains a significant problem. Addressing this challenge requires tools that can quantitatively determine the specificities of both individual proteins and multimeric complexes in an unbiased and comprehensive way. One approach utilizes in vitro selection, high-throughput sequencing, and sequence-specificity landscapes (SEQRS). We outline a SEQRS experiment focused on obtaining the specificity of a multi-protein complex between Drosophila RBPs Pumilio (Pum) and Nanos (Nos). We discuss the necessary controls in this type of experiment and examine how the resulting data can be complemented with structural and cell-based reporter assays. Additionally, SEQRS data can be integrated with functional genomics data to uncover biological function. Finally, we propose extensions of the technique that will enhance our understanding of multi-protein regulatory complexes assembled onto RNA.


Subject(s)
Drosophila Proteins/genetics , RNA-Binding Proteins/genetics , RNA/chemistry , SELEX Aptamer Technique , Sequence Analysis, RNA/methods , Animals , Base Sequence , Binding Sites , DNA Primers/chemistry , DNA Primers/metabolism , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Library , High-Throughput Nucleotide Sequencing , Protein Binding , RNA/genetics , RNA/metabolism , RNA-Binding Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
15.
Elife ; 52016 08 02.
Article in English | MEDLINE | ID: mdl-27482653

ABSTRACT

Collaboration among the multitude of RNA-binding proteins (RBPs) is ubiquitous, yet our understanding of these key regulatory complexes has been limited to single RBPs. We investigated combinatorial translational regulation by Drosophila Pumilio (Pum) and Nanos (Nos), which control development, fertility, and neuronal functions. Our results show how the specificity of one RBP (Pum) is modulated by cooperative RNA recognition with a second RBP (Nos) to synergistically repress mRNAs. Crystal structures of Nos-Pum-RNA complexes reveal that Nos embraces Pum and RNA, contributes sequence-specific contacts, and increases Pum RNA-binding affinity. Nos shifts the recognition sequence and promotes repression complex formation on mRNAs that are not stably bound by Pum alone, explaining the preponderance of sub-optimal Pum sites regulated in vivo. Our results illuminate the molecular mechanism of a regulatory switch controlling crucial gene expression programs, and provide a framework for understanding how the partnering of RBPs evokes changes in binding specificity that underlie regulatory network dynamics.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/physiology , Gene Expression Regulation , RNA-Binding Proteins/metabolism , RNA/metabolism , Repressor Proteins/metabolism , Animals , Crystallography, X-Ray , Drosophila Proteins/chemistry , Models, Molecular , Protein Binding , Protein Conformation , RNA/chemistry , RNA-Binding Proteins/chemistry , Repressor Proteins/chemistry
16.
Trends Biochem Sci ; 41(9): 734-736, 2016 09.
Article in English | MEDLINE | ID: mdl-27427309

ABSTRACT

While a variety of powerful tools exists for analyzing RNA structure, identifying long-range and intermolecular base-pairing interactions has remained challenging. Recently, three groups introduced a high-throughput strategy that uses psoralen-mediated crosslinking to directly identify RNA-RNA duplexes in cells. Initial application of these methods highlights the preponderance of long-range structures within and between RNA molecules and their widespread structural dynamics.


Subject(s)
Base Pairing , RNA/analysis , RNA/chemistry , Thermodynamics
17.
RNA ; 20(8): 1298-319, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24942623

ABSTRACT

PUF proteins are potent repressors that serve important roles in stem cell maintenance, neurological processes, and embryonic development. These functions are driven by PUF protein recognition of specific binding sites within the 3' untranslated regions of target mRNAs. In this study, we investigated mechanisms of repression by the founding PUF, Drosophila Pumilio, and its human orthologs. Here, we evaluated a previously proposed model wherein the Pumilio RNA binding domain (RBD) binds Argonaute, which in turn blocks the translational activity of the eukaryotic elongation factor 1A. Surprisingly, we found that Argonautes are not necessary for repression elicited by Drosophila and human PUFs in vivo. A second model proposed that the RBD of Pumilio represses by recruiting deadenylases to shorten the mRNA's polyadenosine tail. Indeed, the RBD binds to the Pop2 deadenylase and accelerates deadenylation; however, this activity is not crucial for regulation. Rather, we determined that the poly(A) is necessary for repression by the RBD. Our results reveal that poly(A)-dependent repression by the RBD requires the poly(A) binding protein, pAbp. Furthermore, we show that repression by the human PUM2 RBD requires the pAbp ortholog, PABPC1. Pumilio associates with pAbp but does not disrupt binding of pAbp to the mRNA. Taken together, our data support a model wherein the Pumilio RBD antagonizes the ability of pAbp to promote translation. Thus, the conserved function of the PUF RBD is to bind specific mRNAs, antagonize pAbp function, and promote deadenylation.


Subject(s)
Poly A/metabolism , Polyadenylation/physiology , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Argonaute Proteins/chemistry , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Binding Sites , Drosophila Proteins/metabolism , Exoribonucleases/metabolism , Humans , Molecular Sequence Data , Mutation , Nucleotide Motifs , Peptide Elongation Factor 1/metabolism , Protein Binding , Protein Interaction Domains and Motifs , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Sequence Alignment , Ubiquitin-Specific Proteases/metabolism
18.
J Biol Chem ; 287(43): 36370-83, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-22955276

ABSTRACT

PUF proteins are a conserved family of eukaryotic RNA-binding proteins that regulate specific mRNAs: they control many processes including stem cell proliferation, fertility, and memory formation. PUFs repress protein expression from their target mRNAs but the mechanism by which they do so remains unclear, especially for humans. Humans possess two PUF proteins, PUM1 and PUM2, which exhibit similar RNA binding specificities. Here we report new insights into their regulatory activities and mechanisms of action. We developed functional assays to measure sequence-specific repression by PUM1 and PUM2. Both robustly inhibit translation and promote mRNA degradation. Purified PUM complexes were found to contain subunits of the CCR4-NOT (CNOT) complex, which contains multiple enzymes that catalyze mRNA deadenylation. PUMs interact with the CNOT deadenylase subunits in vitro. We used three approaches to determine the importance of deadenylases for PUM repression. First, dominant-negative mutants of CNOT7 and CNOT8 reduced PUM repression. Second, RNA interference depletion of the deadenylases alleviated PUM repression. Third, the poly(A) tail was necessary for maximal PUM repression. These findings demonstrate a conserved mechanism of PUF-mediated repression via direct recruitment of the CCR4-POP2-NOT deadenylase leading to translational inhibition and mRNA degradation. A second, deadenylation independent mechanism was revealed by the finding that PUMs repress an mRNA that lacks a poly(A) tail. Thus, human PUMs are repressors capable of deadenylation-dependent and -independent modes of repression.


Subject(s)
Exoribonucleases/metabolism , Multienzyme Complexes/metabolism , Protein Biosynthesis/physiology , RNA Stability/physiology , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Biological Assay , Exoribonucleases/genetics , HEK293 Cells , Humans , Multienzyme Complexes/genetics , Poly A/genetics , Poly A/metabolism , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Repressor Proteins , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Mol Cell Biol ; 32(2): 527-40, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22064486

ABSTRACT

Drosophila melanogaster Pumilio is an RNA-binding protein that potently represses specific mRNAs. In developing embryos, Pumilio regulates a key morphogen, Hunchback, in collaboration with the cofactor Nanos. To investigate repression by Pumilio and Nanos, we created cell-based assays and found that Pumilio inhibits translation and enhances mRNA decay independent of Nanos. Nanos robustly stimulates repression through interactions with the Pumilio RNA-binding domain. We programmed Pumilio to recognize a new binding site, which garners repression of new target mRNAs. We show that cofactors Brain Tumor and eIF4E Homologous Protein are not obligatory for Pumilio and Nanos activity. The conserved RNA-binding domain of Pumilio was thought to be sufficient for its function. Instead, we demonstrate that three unique domains in the N terminus of Pumilio possess the major repressive activity and can function autonomously. The N termini of insect and vertebrate Pumilio and Fem-3 binding factors (PUFs) are related, and we show that corresponding regions of human PUM1 and PUM2 have repressive activity. Other PUF proteins lack these repression domains. Our findings suggest that PUF proteins have evolved new regulatory functions through protein sequences appended to their conserved PUF repeat RNA-binding domains.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Humans , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...