Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Development ; 150(2)2023 01 15.
Article in English | MEDLINE | ID: mdl-36715566

ABSTRACT

A hallmark of all germ cells is the presence of germ granules: assemblies of proteins and RNA that lack a delineating membrane and are proposed to form via condensation. Germ granules across organisms share several conserved components, including factors required for germ cell fate determination and maintenance, and are thought to be linked to germ cell development. The molecular functions of germ granules, however, remain incompletely understood. In this Development at a Glance article, we survey germ granules across organisms and developmental stages, and highlight emerging themes regarding granule regulation, dynamics and proposed functions.


Subject(s)
Caenorhabditis elegans , Germ Cell Ribonucleoprotein Granules , Animals , Caenorhabditis elegans/metabolism , Germ Cells/metabolism , RNA-Binding Proteins/metabolism , RNA/metabolism , Cytoplasmic Granules/metabolism
2.
Science ; 373(6560): 1218-1224, 2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34516789

ABSTRACT

Biomolecular condensates are cellular compartments that can form by phase separation in the absence of limiting membranes. Studying the P granules of Caenorhabditis elegans, we find that condensate dynamics are regulated by protein clusters that adsorb to the condensate interface. Using in vitro reconstitution, live observations, and theory, we demonstrate that localized assembly of P granules is controlled by MEG-3, an intrinsically disordered protein that forms low dynamic assemblies on P granules. Following classic Pickering emulsion theory, MEG-3 clusters lower surface tension and slow down coarsening. During zygote polarization, MEG-3 recruits the DYRK family kinase MBK-2 to accelerate spatially regulated growth of the P granule emulsion. By tuning condensate-cytoplasm exchange, interfacial clusters regulate the structural integrity of biomolecular condensates, reminiscent of the role of lipid bilayers in membrane-bound organelles.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , Intrinsically Disordered Proteins/metabolism , Animals , Caenorhabditis elegans/metabolism , Oocytes/metabolism , Protein-Tyrosine Kinases/metabolism , RNA-Binding Proteins/metabolism , Zygote/metabolism
3.
Curr Protoc Mol Biol ; 129(1): e102, 2019 12.
Article in English | MEDLINE | ID: mdl-31710422

ABSTRACT

Tagging proteins with fluorescent reporters such as green fluorescent protein (GFP) is a powerful method to determine protein localization, especially when proteins are tagged in the endogenous context to preserve native genomic regulation. However, insertion of fluorescent reporters into the genomes of mammalian cells has required the construction of plasmids containing selection markers and/or extended sequences homologous to the site of insertion (homology arms). Here we describe a streamlined protocol that eliminates all cloning steps by taking advantage of the high propensity of linear DNAs to engage in homology-directed repair of DNA breaks induced by the Cas9 RNA-guided endonuclease. The protocol uses PCR amplicons, or synthetic gene fragments, with short homology arms (30-40 bp) to insert fluorescent reporters at specific genomic locations. The linear DNAs are introduced into cells with preassembled Cas9-crRNA-tracrRNA complexes using one of two transfection procedures, nucleofection or lipofection. The protocol can be completed under a week, with efficiencies ranging from 0.5% to 20% of transfected cells depending on the locus targeted. © 2019 The Authors.


Subject(s)
CRISPR-Cas Systems , DNA, Single-Stranded/genetics , Gene Editing/methods , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Proteins/genetics
4.
Dev Cell ; 50(6): 716-728.e6, 2019 09 23.
Article in English | MEDLINE | ID: mdl-31402283

ABSTRACT

P granules are perinuclear condensates in C. elegans germ cells proposed to serve as hubs for self/non-self RNA discrimination by Argonautes. We report that a mutant (meg-3 meg-4) that does not assemble P granules in primordial germ cells loses competence for RNA-interference over several generations and accumulates silencing small RNAs against hundreds of endogenous genes, including the RNA-interference genes rde-11 and sid-1. In wild type, rde-11 and sid-1 transcripts are heavily targeted by piRNAs and accumulate in P granules but maintain expression. In the primordial germ cells of meg-3 meg-4 mutants, rde-11 and sid-1 transcripts disperse in the cytoplasm with the small RNA biogenesis machinery, become hyper-targeted by secondary sRNAs, and are eventually silenced. Silencing requires the PIWI-class Argonaute PRG-1 and the nuclear Argonaute HRDE-1 that maintains trans-generational silencing of piRNA targets. These observations support a "safe harbor" model for P granules in protecting germline transcripts from piRNA-initiated silencing.


Subject(s)
Caenorhabditis elegans/genetics , Cytoplasmic Granules/genetics , RNA Interference , RNA, Small Interfering/genetics , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins/metabolism , Cell Nucleus/metabolism , Embryo, Nonmammalian/metabolism , Epigenesis, Genetic , Genes, Helminth , Genetic Loci , Germ Cells/metabolism , Models, Biological , Mutation/genetics , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Transcription, Genetic , Up-Regulation/genetics
5.
Development ; 146(6)2019 03 25.
Article in English | MEDLINE | ID: mdl-30814118

ABSTRACT

The MARK/PAR-1 family of kinases are conserved regulators of cell polarity that share a conserved C-terminal kinase-associated domain (KA1). Localization of MARK/PAR-1 kinases to specific regions of the cell cortex is a hallmark of polarized cells. In Caenorhabditiselegans zygotes, PAR-1 localizes to the posterior cortex under the influence of another polarity kinase, aPKC/PKC-3. Here, we report that asymmetric localization of PAR-1 protein is not essential, and that PAR-1 kinase activity is regulated spatially. We find that, as in human MARK1, the PAR-1 KA1 domain is an auto-inhibitory domain that suppresses kinase activity. Auto-inhibition by the KA1 domain functions in parallel with phosphorylation by PKC-3 to suppress PAR-1 activity in the anterior cytoplasm. The KA1 domain also plays an additional role that is essential for germ plasm maintenance and fertility. Our findings suggest that modular regulation of kinase activity by redundant inhibitory inputs contributes to robust symmetry breaking by MARK/PAR-1 kinases in diverse cell types.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Cell Polarity , Gene Expression Regulation, Developmental , Protein Serine-Threonine Kinases/physiology , Alleles , Animals , Cell Lineage , Cytoplasm/metabolism , Green Fluorescent Proteins , Humans , Microscopy, Confocal , Mutation , Phosphorylation , Protein Domains , Protein Kinase C/physiology , RNA Interference , Threonine/chemistry
7.
Proc Natl Acad Sci U S A ; 114(50): E10745-E10754, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29183983

ABSTRACT

The RNA-guided DNA endonuclease Cas9 has emerged as a powerful tool for genome engineering. Cas9 creates targeted double-stranded breaks (DSBs) in the genome. Knockin of specific mutations (precision genome editing) requires homology-directed repair (HDR) of the DSB by synthetic donor DNAs containing the desired edits, but HDR has been reported to be variably efficient. Here, we report that linear DNAs (single and double stranded) engage in a high-efficiency HDR mechanism that requires only ∼35 nucleotides of homology with the targeted locus to introduce edits ranging from 1 to 1,000 nucleotides. We demonstrate the utility of linear donors by introducing fluorescent protein tags in human cells and mouse embryos using PCR fragments. We find that repair is local, polarity sensitive, and prone to template switching, characteristics that are consistent with gene conversion by synthesis-dependent strand annealing. Our findings enable rational design of synthetic donor DNAs for efficient genome editing.


Subject(s)
Bacterial Proteins/metabolism , DNA Repair , Endonucleases/metabolism , Gene Editing/methods , Animals , CRISPR-Associated Protein 9 , CRISPR-Cas Systems , DNA Breaks, Double-Stranded , HEK293 Cells , Humans , Mice , Polymerase Chain Reaction , Sequence Homology, Nucleic Acid
8.
Methods ; 121-122: 86-93, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28392263

ABSTRACT

The ability to introduce targeted edits in the genome of model organisms is revolutionizing the field of genetics. State-of-the-art methods for precision genome editing use RNA-guided endonucleases to create double-strand breaks (DSBs) and DNA templates containing the edits to repair the DSBs. Following this strategy, we have developed a protocol to create precise edits in the C. elegans genome. The protocol takes advantage of two innovations to improve editing efficiency: direct injection of CRISPR-Cas9 ribonucleoprotein complexes and use of linear DNAs with short homology arms as repair templates. The protocol requires no cloning or selection, and can be used to generate base and gene-size edits in just 4days. Point mutations, insertions, deletions and gene replacements can all be created using the same experimental pipeline.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Cas Systems , Endonucleases/genetics , Gene Editing/methods , Gene Knock-In Techniques , Gene Transfer Techniques , RNA, Guide, Kinetoplastida/genetics , Alleles , Animals , Animals, Genetically Modified , Bacterial Proteins/metabolism , Base Sequence , CRISPR-Associated Protein 9 , Caenorhabditis elegans/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , DNA/genetics , DNA/metabolism , DNA Breaks, Double-Stranded , Embryo, Nonmammalian , Endonucleases/metabolism , Gene Targeting/methods , Genome , Microinjections , Mutation , RNA, Guide, Kinetoplastida/metabolism , Recombinational DNA Repair
9.
Genetics ; 201(1): 47-54, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26187122

ABSTRACT

Homology-directed repair (HDR) of breaks induced by the RNA-programmed nuclease Cas9 has become a popular method for genome editing in several organisms. Most HDR protocols rely on plasmid-based expression of Cas9 and the gene-specific guide RNAs. Here we report that direct injection of in vitro-assembled Cas9-CRISPR RNA (crRNA) trans-activating crRNA (tracrRNA) ribonucleoprotein complexes into the gonad of Caenorhabditis elegans yields HDR edits at a high frequency. Building on our earlier finding that PCR fragments with 35-base homology are efficient repair templates, we developed an entirely cloning-free protocol for the generation of seamless HDR edits without selection. Combined with the co-CRISPR method, this protocol is sufficiently robust for use with low-efficiency guide RNAs and to generate complex edits, including ORF replacement and simultaneous tagging of two genes with fluorescent proteins.


Subject(s)
CRISPR-Cas Systems , Caenorhabditis elegans/genetics , Genetic Engineering/methods , Ribonucleoproteins/administration & dosage , Animals , DNA Repair , Injections , Organisms, Genetically Modified , RNA, Guide, Kinetoplastida/genetics
10.
Mol Biol Cell ; 26(8): 1476-90, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25694449

ABSTRACT

When eukaryotic cells respond to stress, gene expression pathways change to selectively export and translate subsets of mRNAs. Translationally repressed mRNAs accumulate in cytoplasmic foci known as stress granules (SGs). SGs are in dynamic equilibrium with the translational machinery, but mechanisms controlling this are unclear. Gle1 is required for DEAD-box protein function during mRNA export and translation. We document that human Gle1 (hGle1) is a critical regulator of translation during stress. hGle1 is recruited to SGs, and hGLE1 small interfering RNA-mediated knockdown perturbs SG assembly, resulting in increased numbers of smaller SGs. The rate of SG disassembly is also delayed. Furthermore, SG hGle1-depletion defects correlate with translation perturbations, and the hGle1 role in SGs is independent of mRNA export. Interestingly, we observe isoform-specific roles for hGle1 in which SG function requires hGle1A, whereas mRNA export requires hGle1B. We find that the SG defects in hGle1-depleted cells are rescued by puromycin or DDX3 expression. Together with recent links of hGLE1 mutations in amyotrophic lateral sclerosis patients, these results uncover a paradigm for hGle1A modulating the balance between translation and SGs during stress and disease.


Subject(s)
Cytoplasmic Granules/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Protein Biosynthesis/physiology , Stress, Physiological , Cell Line , Down-Regulation , Humans , Nucleocytoplasmic Transport Proteins/genetics , Protein Isoforms/metabolism
11.
Hum Mol Genet ; 24(5): 1363-73, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25343993

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterized by the selective death of motor neurons. Causative mutations in the global RNA-processing proteins TDP-43 and FUS among others, as well as their aggregation in ALS patients, have identified defects in RNA metabolism as an important feature in this disease. Lethal congenital contracture syndrome 1 and lethal arthrogryposis with anterior horn cell disease are autosomal recessive fetal motor neuron diseases that are caused by mutations in another global RNA-processing protein, hGle1. In this study, we carried out the first screening of GLE1 in ALS patients (173 familial and 760 sporadic) and identified 2 deleterious mutations (1 splice site and 1 nonsense mutation) and 1 missense mutation. Functional analysis of the deleterious mutants revealed them to be unable to rescue motor neuron pathology in zebrafish morphants lacking Gle1. Furthermore, in HeLa cells, both mutations caused a depletion of hGle1 at the nuclear pore where it carries out an essential role in nuclear export of mRNA. These results suggest a haploinsufficiency mechanism and point to a causative role for GLE1 mutations in ALS patients. This further supports the involvement of global defects in RNA metabolism in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Codon, Nonsense , Mutation, Missense , Nucleocytoplasmic Transport Proteins/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Arthrogryposis/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Haploinsufficiency/genetics , HeLa Cells , Humans , Microscopy, Confocal , Motor Neurons/pathology , Nuclear Pore/genetics , Nuclear Pore/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Pedigree , Protein Processing, Post-Translational , RNA Splicing , RNA, Messenger/metabolism , Zebrafish
12.
Adv Biol Regul ; 54: 74-91, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24275432

ABSTRACT

A critical step during gene expression is the directional export of nuclear messenger (m)RNA through nuclear pore complexes (NPCs) to the cytoplasm. During export, Gle1 in conjunction with inositol hexakisphosphate (IP6) spatially regulates the activity of the DEAD-box protein Dbp5 at the NPC cytoplasmic face. GLE1 mutations are causally linked to the human diseases lethal congenital contracture syndrome 1 (LCCS-1) and lethal arthrogryposis with anterior horn cell disease (LAAHD). Here, structure prediction and functional analysis provide strong evidence to suggest that the LCCS-1 and LAAHD disease mutations disrupt the function of Gle1 in mRNA export. Strikingly, direct fluorescence microscopy in living cells reveals a dramatic loss of steady-state NPC localization for GFP-gle1 proteins expressed from human gle1 genes harboring LAAHD and LCCS-1 mutations. The potential significance of these residues is further clarified by analyses of sequence and predicted structural conservation. This work offers insights into the perturbed mechanisms underlying human LCCS-1 and LAAHD disease states and emphasizes the potential impact of altered mRNA transport and gene expression in human disease.


Subject(s)
Arthrogryposis/metabolism , Cell Nucleus/metabolism , Motor Neuron Disease/metabolism , Nucleocytoplasmic Transport Proteins/chemistry , Nucleocytoplasmic Transport Proteins/metabolism , RNA, Messenger/metabolism , Animals , Arthrogryposis/genetics , Cell Nucleus/genetics , Humans , Motor Neuron Disease/genetics , Nucleocytoplasmic Transport Proteins/genetics , RNA Transport , RNA, Messenger/genetics
13.
Cell ; 155(3): 582-93, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24243016

ABSTRACT

The conserved multifunctional protein Gle1 regulates gene expression at multiple steps: nuclear mRNA export, translation initiation, and translation termination. A GLE1 mutation (FinMajor) is causally linked to human lethal congenital contracture syndrome-1 (LCCS1); however, the resulting perturbations on Gle1 molecular function were unknown. FinMajor results in a proline-phenylalanine-glutamine peptide insertion within the uncharacterized Gle1 coiled-coil domain. Here, we find that Gle1 self-associates both in vitro and in living cells via the coiled-coil domain. Electron microscopy reveals that high-molecular-mass Gle1 oligomers form ?26 nm diameter disk-shaped particles. With the Gle1-FinMajor protein, these particles are malformed. Moreover, functional assays document a specific requirement for proper Gle1 oligomerization during mRNA export, but not for Gle1's roles in translation. These results identify a mechanistic step in Gle1's mRNA export function at nuclear pore complexes and directly implicate altered export in LCCS1 disease pathology.


Subject(s)
Arthrogryposis/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , RNA, Messenger/metabolism , Active Transport, Cell Nucleus , Arthrogryposis/genetics , Arthrogryposis/pathology , HeLa Cells , Humans , Mutation , Nuclear Pore/metabolism , Saccharomyces cerevisiae/metabolism
14.
Nucleus ; 2(6): 540-8, 2011.
Article in English | MEDLINE | ID: mdl-22064466

ABSTRACT

Gene expression is a stepwise process involving distinct cellular processes including transcription, mRNA (mRNA) processing, mRNA export, and translation. As mRNAs are being synthesized, proteins associate with the RNA to form messenger ribonucleoprotein particles (mRNPs). Previous studies have demonstrated that the RNA-binding protein composition of these mRNPs is dynamic, changing as the mRNP moves through the different steps of gene expression, and playing a critical role in these events. An important step during this maturation process occurs at the cytoplasmic face of the nuclear pore complex (NPC) where the export protein Gle1 bound to inositol hexakisphosphate (IP 6) spatially activates the ATP-hydrolysis and mRNP-remodeling activity of the DEAD-box protein Dbp5. Recent work from our laboratory and others has provided important insights into the function and regulation of Dbp5. These include a more detailed explanation of the mechanism of Dbp5 RNP remodeling, the role of Gle1-IP6 in stimulating Dbp5 ATPase activity, and the identification of a novel paradigm for regulation of Dbp5 by Nup159. Based on in vitro biochemical assays, X-ray crystallography, and corresponding in vivo phenotypes, we propose here an updated model of the Dbp5 cycle during mRNP export through the NPC. This takes into account all available data and provides a platform for future studies.


Subject(s)
Cell Nucleus/metabolism , DEAD-box RNA Helicases/metabolism , Models, Biological , Nuclear Pore Complex Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Phytic Acid/metabolism , Ribonucleoproteins/metabolism , Active Transport, Cell Nucleus/physiology , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Animals , Cell Nucleus/genetics , Cytoplasm/genetics , Cytoplasm/metabolism , DEAD-box RNA Helicases/genetics , Gene Expression Regulation/physiology , Humans , Nuclear Pore Complex Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics , Ribonucleoproteins/genetics
15.
Genes Dev ; 25(10): 1052-64, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21576265

ABSTRACT

Nuclear export of messenger RNA (mRNA) occurs by translocation of mRNA/protein complexes (mRNPs) through nuclear pore complexes (NPCs). The DEAD-box protein Dbp5 mediates export by triggering removal of mRNP proteins in a spatially controlled manner. This requires Dbp5 interaction with Nup159 in NPC cytoplasmic filaments and activation of Dbp5's ATPase activity by Gle1 bound to inositol hexakisphosphate (IP(6)). However, the precise sequence of events within this mechanism has not been fully defined. Here we analyze dbp5 mutants that alter ATP binding, ATP hydrolysis, or RNA binding. We found that ATP binding and hydrolysis are required for efficient Dbp5 association with NPCs. Interestingly, mutants defective for RNA binding are dominant-negative (DN) for mRNA export in yeast and human cells. We show that the DN phenotype stems from competition with wild-type Dbp5 for Gle1 at NPCs. The Dbp5-Gle1 interaction is limiting for export and, importantly, can be independent of Nup159. Fluorescence recovery after photobleaching experiments in yeast show a very dynamic association between Dbp5 and NPCs, averaging <1 sec, similar to reported NPC translocation rates for mRNPs. This work reveals critical steps in the Gle1-IP(6)/Dbp5/Nup159 cycle, and suggests that the number of remodeling events mediated by a single Dbp5 is limited.


Subject(s)
Cell Nucleus/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Nuclear Pore/metabolism , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Active Transport, Cell Nucleus , Adenosine Triphosphate/metabolism , Cell Line, Tumor , HeLa Cells , Humans , Hydrolysis , Mutation , Nuclear Pore Complex Proteins/metabolism , Phenotype , Protein Binding/genetics , Saccharomyces cerevisiae/growth & development
16.
Mol Cancer Res ; 9(6): 712-23, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21460181

ABSTRACT

Metastatic melanoma is an aggressive skin disease for which there are no effective therapies. Emerging evidence indicates that melanomas can be sensitized to chemotherapy by increasing integrin function. Current integrin therapies work by targeting the extracellular domain, resulting in complete gains or losses of integrin function that lead to mechanism-based toxicities. An attractive alternative approach is to target proteins, such as vinculin, that associate with the integrin cytoplasmic domains and regulate its ligand-binding properties. Here, we report that a novel reagent, denoted vinculin-activating peptide or VAP, increases integrin activity from within the cell, as measured by elevated (i) numbers of active integrins, (ii) adhesion of cells to extracellular matrix ligands, (iii) numbers of cell-matrix adhesions, and (iv) downstream signaling. These effects are dependent on both integrins and a key regulatory residue A50 in the vinculin head domain. We further show that VAP dramatically increases the sensitivity of melanomas to chemotherapy in clonal growth assays and in vivo mouse models of melanoma. Finally, we show that the increase in chemosensitivity results from increases in DNA damage-induced apoptosis in a p53-dependent manner. Collectively, these findings show that integrin function can be manipulated from within the cell and validate integrins as a new therapeutic target for the treatment of chemoresistant melanomas.


Subject(s)
Antineoplastic Agents/pharmacology , Bacterial Proteins/pharmacology , Drug Resistance, Neoplasm/drug effects , Integrins/metabolism , Melanoma/metabolism , Peptide Fragments/pharmacology , Skin Neoplasms/metabolism , Vinculin/agonists , Animals , Antineoplastic Agents/therapeutic use , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Line, Tumor , Fibroblasts/metabolism , HeLa Cells , Humans , Melanoma/drug therapy , Mice , Skin Neoplasms/drug therapy
17.
Nat Cell Biol ; 11(9): 1069-80, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19701196

ABSTRACT

Microtubules are indispensable for Golgi complex assembly and maintenance, which are integral parts of cytoplasm organization during interphase in mammalian cells. Here, we show that two discrete microtubule subsets drive two distinct, yet simultaneous, stages of Golgi assembly. In addition to the radial centrosomal microtubule array, which positions the Golgi in the centre of the cell, we have identified a role for microtubules that form at the Golgi membranes in a manner dependent on the microtubule regulators CLASPs. These Golgi-derived microtubules draw Golgi ministacks together in tangential fashion and are crucial for establishing continuity and proper morphology of the Golgi complex. We propose that specialized functions of these two microtubule arrays arise from their specific geometries. Further, we demonstrate that directional post-Golgi trafficking and cell migration depend on Golgi-associated CLASPs, suggesting that correct organization of the Golgi complex by microtubules is essential for cell polarization and motility.


Subject(s)
Cell Movement , Cell Polarity , Golgi Apparatus/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Biological Transport , Cell Line , Centrosome/metabolism , Dyneins/metabolism , Humans , Mitosis
18.
Cell ; 134(4): 624-33, 2008 Aug 22.
Article in English | MEDLINE | ID: mdl-18724935

ABSTRACT

Gene expression requires proper messenger RNA (mRNA) export and translation. However, the functional links between these consecutive steps have not been fully defined. Gle1 is an essential, conserved mRNA export factor whose export function is dependent on the small molecule inositol hexakisphosphate (IP(6)). Here, we show that both Gle1 and IP(6) are required for efficient translation termination in Saccharomyces cerevisiae and that Gle1 interacts with termination factors. In addition, Gle1 has a conserved physical association with the initiation factor eIF3, and gle1 mutants display genetic interactions with the eIF3 mutant nip1-1. Strikingly, gle1 mutants have defects in initiation, whereas strains lacking IP(6) do not. We propose that Gle1 functions together with IP(6) and the DEAD-box protein Dbp5 to regulate termination. However, Gle1 also independently mediates initiation. Thus, Gle1 is uniquely positioned to coordinate the mRNA export and translation mechanisms. These results directly impact models for perturbation of Gle1 function in pathophysiology.


Subject(s)
Carrier Proteins/metabolism , Phytic Acid/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , DEAD-box RNA Helicases/metabolism , Eukaryotic Initiation Factor-3/metabolism , Nuclear Pore Complex Proteins , Nucleocytoplasmic Transport Proteins/metabolism , Peptide Termination Factors/metabolism , Protein Isoforms/metabolism , Saccharomyces cerevisiae/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...