Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 2668, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33976151

ABSTRACT

Telomeres are bound by dedicated proteins, which protect them from DNA damage and regulate telomere length homeostasis. In the nematode Caenorhabditis elegans, a comprehensive understanding of the proteins interacting with the telomere sequence is lacking. Here, we harnessed a quantitative proteomics approach to identify TEBP-1 and TEBP-2, two paralogs expressed in the germline and embryogenesis that associate to telomeres in vitro and in vivo. tebp-1 and tebp-2 mutants display strikingly distinct phenotypes: tebp-1 mutants have longer telomeres than wild-type animals, while tebp-2 mutants display shorter telomeres and a Mortal Germline. Notably, tebp-1;tebp-2 double mutant animals have synthetic sterility, with germlines showing signs of severe mitotic and meiotic arrest. Furthermore, we show that POT-1 forms a telomeric complex with TEBP-1 and TEBP-2, which bridges TEBP-1/-2 with POT-2/MRT-1. These results provide insights into the composition and organization of a telomeric protein complex in C. elegans.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , DNA-Binding Proteins/metabolism , DNA/metabolism , Multiprotein Complexes/metabolism , Telomere-Binding Proteins/metabolism , Telomere/metabolism , Animals , Animals, Genetically Modified , Base Sequence , Binding Sites/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/classification , Caenorhabditis elegans Proteins/genetics , DNA/genetics , DNA-Binding Proteins/genetics , Germ Cells/metabolism , Microscopy, Fluorescence/methods , Multiprotein Complexes/genetics , Mutation , Phylogeny , Protein Binding , Protein Isoforms/classification , Protein Isoforms/genetics , Protein Isoforms/metabolism , Telomere/genetics , Telomere-Binding Proteins/classification , Telomere-Binding Proteins/genetics
2.
PLoS Genet ; 13(3): e1006614, 2017 03.
Article in English | MEDLINE | ID: mdl-28301465

ABSTRACT

Structural Maintenance of Chromosomes (SMC) family proteins participate in multisubunit complexes that govern chromosome structure and dynamics. SMC-containing condensin complexes create chromosome topologies essential for mitosis/meiosis, gene expression, recombination, and repair. Many eukaryotes have two condensin complexes (I and II); C. elegans has three (I, II, and the X-chromosome specialized condensin IDC) and their regulation is poorly understood. Here we identify a novel SMC-like protein, SMCL-1, that binds to C. elegans condensin SMC subunits, and modulates condensin functions. Consistent with a possible role as a negative regulator, loss of SMCL-1 partially rescued the lethal and sterile phenotypes of a hypomorphic condensin mutant, while over-expression of SMCL-1 caused lethality, chromosome mis-segregation, and disruption of condensin IDC localization on X chromosomes. Unlike canonical SMC proteins, SMCL-1 lacks hinge and coil domains, and its ATPase domain lacks conserved amino acids required for ATP hydrolysis, leading to the speculation that it may inhibit condensin ATPase activity. SMCL-1 homologs are apparent only in the subset of Caenorhabditis species in which the condensin I and II subunit SMC-4 duplicated to create the condensin IDC- specific subunit DPY-27, suggesting that SMCL-1 helps this lineage cope with the regulatory challenges imposed by evolution of a third condensin complex. Our findings uncover a new regulator of condensins and highlight how the duplication and divergence of SMC complex components in various lineages has created new proteins with diverse functions in chromosome dynamics.


Subject(s)
Adenosine Triphosphatases/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , DNA-Binding Proteins/genetics , Multiprotein Complexes/genetics , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Caenorhabditis elegans/embryology , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/classification , Caenorhabditis elegans Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Meiosis/genetics , Microscopy, Confocal , Mitosis/genetics , Multiprotein Complexes/metabolism , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phylogeny , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism , Sequence Homology, Amino Acid , X Chromosome/genetics
3.
Ultramicroscopy ; 164: 46-50, 2016 May.
Article in English | MEDLINE | ID: mdl-27010412

ABSTRACT

An alternative method has been assessed; diffraction patterns derived from the single particle data set were used to perform the first round of classification in creating the initial averages for proteins data with symmetrical morphology. The test protein set was a collection of Caenorhabditis elegans small heat shock protein 17 obtained by Cryo EM, which has a tetrahedral (12-fold) symmetry. It is demonstrated that the initial classification on diffraction patterns is workable as well as the real-space classification that is based on the phase contrast. The test results show that the information from diffraction patterns has the enough details to make the initial model faithful. The potential advantage using the alternative method is twofold, the ability to handle the sets with poor signal/noise or/and that break the symmetry properties.


Subject(s)
Caenorhabditis elegans Proteins/classification , Heat-Shock Proteins, Small/classification , Microscopy, Electron, Transmission , Animals , Caenorhabditis elegans/chemistry , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/ultrastructure , Cryoelectron Microscopy , Crystallization , Heat-Shock Proteins, Small/chemistry , Heat-Shock Proteins, Small/ultrastructure , Image Processing, Computer-Assisted , Imaging, Three-Dimensional
4.
Proteins ; 84(3): 397-407, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26757344

ABSTRACT

Histone post-translational modifications play a critical role in the regulation of gene expression. Methylation of lysines at N-terminal tails of histones has been shown to be involved in such regulation. While this modification was long considered to be irreversible, two different classes of enzymes capable of carrying out the demethylation of histone lysines were recently identified: the oxidases, such as LSD1, and the oxygenases (JmjC-containing). Here, a family-wide analysis of the second of these classes is proposed, with over 300 proteins studied at the sequence level. We show that a correlated evolution analysis yields some position/residue pairs which are critical at comparing JmjC sequences and enables the classification of JmjC domains into five families. A few positions appear more frequently among conditions, such as positions 23 (directly C-terminal to the second iron ligand), 24, 252 and 253 (directly N-terminal to a conserved Asn). Implications of family conditions are studied in detail on PHF2, revealing the meaningfulness of the sequence-derived conditions at the structural level. These results should help obtain insights on the diversity of JmjC-containing proteins solely by considering some of the amino acids present in their JmjC domain.


Subject(s)
Histone Demethylases/chemistry , Amino Acid Sequence , Animals , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/classification , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/classification , Catalytic Domain , Fungal Proteins/chemistry , Fungal Proteins/classification , Histone Demethylases/classification , Humans , Methylation , Molecular Sequence Data , Protein Processing, Post-Translational , Sequence Alignment , Sequence Analysis, Protein , Sequence Homology, Amino Acid , Substrate Specificity
5.
Mol Cell ; 58(6): 1067-78, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26009280

ABSTRACT

Small heat shock proteins (sHsps) are ubiquitous molecular chaperones that prevent the aggregation of unfolding proteins during proteotoxic stress. In Caenorhabditis elegans, Sip1 is the only sHsp exclusively expressed in oocytes and embryos. Here, we demonstrate that Sip1 is essential for heat shock survival of reproducing adults and embryos. X-ray crystallography and electron microscopy revealed that Sip1 exists in a range of well-defined globular assemblies consisting of two half-spheres, each made of dimeric "spokes." Strikingly, the oligomeric distribution of Sip1 as well as its chaperone activity depend on pH, with a trend toward smaller species and higher activity at acidic conditions such as present in nematode eggs. The analysis of the interactome shows that Sip1 has a specific substrate spectrum including proteins that are essential for embryo development.


Subject(s)
Caenorhabditis elegans Proteins/chemistry , Heat-Shock Proteins, Small/chemistry , Molecular Chaperones/chemistry , Protein Conformation , Amino Acid Sequence , Animals , Blotting, Western , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/classification , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , Heat-Shock Proteins, Small/genetics , Heat-Shock Proteins, Small/metabolism , Hydrogen-Ion Concentration , Models, Molecular , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Molecular Sequence Data , Mutation , Phylogeny , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Temperature
6.
PLoS One ; 10(2): e0117444, 2015.
Article in English | MEDLINE | ID: mdl-25688864

ABSTRACT

RIO protein kinases (RIOKs) are a relatively conserved family of enzymes implicated in cell cycle control and ribosomal RNA processing. Despite their functional importance, they remain a poorly understood group of kinases in multicellular organisms. Here, we show that the C. elegans genome contains one member of each of the three RIOK sub-families and that each of the genes coding for them has a unique tissue expression pattern. Our analysis showed that the gene encoding RIOK-1 (riok-1) was broadly and strongly expressed. Interestingly, the intestinal expression of riok-1 was dependent upon two putative binding sites for the oxidative and xenobiotic stress response transcription factor SKN-1. RNA interference (RNAi)-mediated knock down of riok-1 resulted in germline defects, including defects in germ line stem cell proliferation, oocyte maturation and the production of endomitotic oocytes. Taken together, our findings indicate new functions for RIOK-1 in post mitotic tissues and in reproduction.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Protein Kinases/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/classification , Cell Proliferation , Female , Genome , Germ Cells/cytology , Germ Cells/metabolism , Gonads/metabolism , Male , Phylogeny , Promoter Regions, Genetic , Protein Kinases/classification , Protein Kinases/genetics , RNA Interference , RNA, Double-Stranded/metabolism , Sequence Alignment
7.
Nucleic Acids Res ; 43(1): 208-24, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25510497

ABSTRACT

As a champion of small RNA research for two decades, Caenorhabditis elegans has revealed the essential Argonaute CSR-1 to play key nuclear roles in modulating chromatin, chromosome segregation and germline gene expression via 22G-small RNAs. Despite CSR-1 being preserved among diverse nematodes, the conservation and divergence in function of the targets of small RNA pathways remains poorly resolved. Here we apply comparative functional genomic analysis between C. elegans and Caenorhabditis briggsae to characterize the CSR-1 pathway, its targets and their evolution. C. briggsae CSR-1-associated small RNAs that we identified by immunoprecipitation-small RNA sequencing overlap with 22G-RNAs depleted in cbr-csr-1 RNAi-treated worms. By comparing 22G-RNAs and target genes between species, we defined a set of CSR-1 target genes with conserved germline expression, enrichment in operons and more slowly evolving coding sequences than other genes, along with a small group of evolutionarily labile targets. We demonstrate that the association of CSR-1 with chromatin is preserved, and show that depletion of cbr-csr-1 leads to chromosome segregation defects and embryonic lethality. This first comparative characterization of a small RNA pathway in Caenorhabditis establishes a conserved nuclear role for CSR-1 and highlights its key role in germline gene regulation across multiple animal species.


Subject(s)
Argonaute Proteins/metabolism , Caenorhabditis/genetics , Helminth Proteins/metabolism , RNA, Small Untranslated/metabolism , Animals , Argonaute Proteins/chemistry , Argonaute Proteins/classification , Caenorhabditis/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/classification , Chromatin/metabolism , Chromosome Segregation , Gene Expression Regulation
8.
Cell Mol Life Sci ; 71(17): 3339-61, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24458371

ABSTRACT

Protein misfolding and aggregation as a consequence of impaired protein homeostasis (proteostasis) not only characterizes numerous age-related diseases but also the aging process itself. Functionally related to the aging process are, among others, ribosomal proteins, suggesting an intimate link between proteostasis and aging. We determined by iTRAQ quantitative proteomic analysis in C. elegans how the proteome changes with age and in response to heat shock. Levels of ribosomal proteins and mitochondrial chaperones were decreased in aged animals, supporting the notion that proteostasis is altered during aging. Mitochondrial enzymes of the tricarboxylic acid cycle and the electron transport chain were also reduced, consistent with an age-associated energy impairment. Moreover, we observed an age-associated decline in the heat shock response. In order to determine how protein synthesis is altered in aging and in response to heat shock, we complemented our global analysis by determining the de novo proteome. For that, we established a novel method that enables both the visualization and identification of de novo synthesized proteins, by incorporating the non-canonical methionine analogue, azidohomoalanine (AHA), into the nascent polypeptides, followed by reacting the azide group of AHA by 'click chemistry' with an alkyne-labeled tag. Our analysis of AHA-tagged peptides demonstrated that the decreased abundance of, for example, ribosomal proteins in aged animals is not solely due to degradation but also reflects a relative decrease in their synthesis. Interestingly, although the net rate of protein synthesis is reduced in aged animals, our analyses indicate that the synthesis of certain proteins such as the vitellogenins increases with age.


Subject(s)
Aging/physiology , Caenorhabditis elegans Proteins/biosynthesis , Heat-Shock Response/physiology , Proteome , Alanine/analogs & derivatives , Alanine/metabolism , Animals , Blotting, Western , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/classification , Caenorhabditis elegans Proteins/genetics , Click Chemistry , Forecasting , Gene Expression Regulation , Genes, Helminth , Heat-Shock Response/genetics , Hot Temperature , Mass Spectrometry , Microscopy, Fluorescence , Protein Folding , Proteostasis Deficiencies/metabolism
9.
PLoS One ; 7(11): e49387, 2012.
Article in English | MEDLINE | ID: mdl-23185324

ABSTRACT

Rab monomeric GTPases regulate specific aspects of vesicle transport in eukaryotes including coat recruitment, uncoating, fission, motility, target selection and fusion. Moreover, individual Rab proteins function at specific sites within the cell, for example the ER, golgi and early endosome. Importantly, the localization and function of individual Rab subfamily members are often conserved underscoring the significant contributions that model organisms such as Caenorhabditis elegans can make towards a better understanding of human disease caused by Rab and vesicle trafficking malfunction. With this in mind, a bioinformatics approach was first taken to identify and classify the complete C. elegans Rab family placing individual Rabs into specific subfamilies based on molecular phylogenetics. For genes that were difficult to classify by sequence similarity alone, we did a comparative analysis of intron position among specific subfamilies from yeast to humans. This two-pronged approach allowed the classification of 30 out of 31 C. elegans Rab proteins identified here including Rab31/Rab50, a likely member of the last eukaryotic common ancestor (LECA). Second, a molecular toolset was created to facilitate research on biological processes that involve Rab proteins. Specifically, we used Gateway-compatible C. elegans ORFeome clones as starting material to create 44 full-length, sequence-verified, dominant-negative (DN) and constitutive active (CA) rab open reading frames (ORFs). Development of this toolset provided independent research projects for students enrolled in a research-based molecular techniques course at California State University, East Bay (CSUEB).


Subject(s)
Caenorhabditis elegans Proteins/classification , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Computational Biology/methods , Multigene Family , rab GTP-Binding Proteins/classification , rab GTP-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans Proteins/chemistry , Clone Cells , Conserved Sequence/genetics , Humans , Introns/genetics , Molecular Sequence Data , Open Reading Frames/genetics , Phylogeny , RNA Splicing/genetics , Reproducibility of Results , Sequence Alignment , rab GTP-Binding Proteins/chemistry
10.
PLoS One ; 7(5): e36770, 2012.
Article in English | MEDLINE | ID: mdl-22701514

ABSTRACT

Calcium (Ca⁺²) is a ubiquitous messenger in eukaryotes including Caenorhabditis. Ca⁺²-mediated signalling processes are usually carried out through well characterized proteins like calmodulin (CaM) and other Ca⁺² binding proteins (CaBP). These proteins interact with different targets and activate it by bringing conformational changes. Majority of the EF-hand proteins in Caenorhabditis contain Ca⁺² binding motifs. Here, we have performed homology modelling of CaM-like proteins using the crystal structure of Drosophila melanogaster CaM as a template. Molecular docking was applied to explore the binding mechanism of CaM-like proteins and IQ1 motif which is a ∼25 residues and conform to the consensus sequence (I, L, V)QXXXRXXXX(R,K) to serve as a binding site for different EF hand proteins. We made an attempt to identify all the EF-hand (a helix-loop-helix structure characterized by a 12 residues loop sequence involved in metal coordination) containing proteins and their Ca⁺² binding affinity in Caenorhabditis by analysing the complete genome sequence. Docking studies revealed that F165, F169, L29, E33, F44, L57, M61, M96, M97, M108, G65, V115, F93, N104, E144 of CaM-like protein is involved in the interaction with IQ1 motif. A maximum of 170 EF-hand proteins and 39 non-EF-hand proteins with Ca⁺²/metal binding motif were identified. Diverse proteins including enzyme, transcription, translation and large number of unknown proteins have one or more putative EF-hands. Phylogenetic analysis revealed seven major classes/groups that contain some families of proteins. Various domains that we identified in the EF-hand proteins (uncharacterized) would help in elucidating their functions. It is the first report of its kind where calcium binding loop sequences of EF-hand proteins were analyzed to decipher their calcium affinities. Variation in Ca⁺²-binding affinity of EF-hand CaBP could be further used to study the behaviour of these proteins. Our analyses postulated that Ca⁺² is likely to be key player in Caenorhabditis cell signalling.


Subject(s)
Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis/metabolism , Calcium-Binding Proteins/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Animals , Caenorhabditis/genetics , Caenorhabditis elegans Proteins/classification , Caenorhabditis elegans Proteins/genetics , Calcium-Binding Proteins/genetics , Drosophila Proteins/genetics , EF Hand Motifs/genetics , EF Hand Motifs/physiology , Phylogeny , Protein Structure, Secondary , Protein Structure, Tertiary , Structural Homology, Protein
11.
PLoS One ; 6(6): e21343, 2011.
Article in English | MEDLINE | ID: mdl-21712952

ABSTRACT

Adiponectin is an adipokine with insulin-sensitising actions in vertebrates. Its receptors, AdipoR1 and AdipoR2, are PAQR-type proteins with 7-transmembrane domains and topologies reversed that of GPCR's, i.e. their C-termini are extracellular. We identified three adiponectin receptor homologs in the nematode C. elegans, named paqr-1, paqr-2 and paqr-3. These are differently expressed in the intestine (the main fat-storing tissue), hypodermis, muscles, neurons and secretory tissues, from which they could exert systemic effects. Analysis of mutants revealed that paqr-1 and -2 are novel metabolic regulators in C. elegans and that they act redundantly but independently from paqr-3. paqr-2 is the most important of the three paqr genes: mutants grow poorly, fail to adapt to growth at low temperature, and have a very high fat content with an abnormal enrichment in long (C20) poly-unsaturated fatty acids when combined with the paqr-1 mutation. paqr-2 mutants are also synthetic lethal with mutations in nhr-49, sbp-1 and fat-6, which are C. elegans homologs of nuclear hormone receptors, SREBP and FAT-6 (a Δ9 desaturase), respectively. Like paqr-2, paqr-1 is also synthetic lethal with sbp-1. Mutations in aak-2, the C. elegans homolog of AMPK, or nhr-80, another nuclear hormone receptor gene, suppress the growth phenotype of paqr-2 mutants, probably because they restore the balance between energy expenditure and storage. We conclude that paqr-1 and paqr-2 are receptors that regulate fatty acid metabolism and cold adaptation in C. elegans, that their main function is to promote energy utilization rather than storage, and that PAQR class proteins have regulated metabolism in metazoans for at least 700 million years.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/physiology , Energy Metabolism/physiology , Homeostasis , Protein Isoforms/metabolism , Receptors, Adiponectin/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/anatomy & histology , Caenorhabditis elegans Proteins/classification , Caenorhabditis elegans Proteins/genetics , Diet , Humans , Lipid Metabolism/genetics , Mutation , Phenotype , Phylogeny , Protein Isoforms/classification , Protein Isoforms/genetics , Receptors, Adiponectin/classification , Receptors, Adiponectin/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
12.
Mol Biol Cell ; 22(14): 2564-78, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21613545

ABSTRACT

In yeast the Golgi-associated retrograde protein (GARP) complex is required for tethering of endosome-derived transport vesicles to the late Golgi. It consists of four subunits--Vps51p, Vps52p, Vps53p, and Vps54p--and shares similarities with other multimeric tethering complexes, such as the conserved oligomeric Golgi (COG) and the exocyst complex. Here we report the functional characterization of the GARP complex in the nematode Caenorhabditis elegans. Furthermore, we identified the C. elegans Vps51 subunit, which is conserved in all eukaryotes. GARP mutants are viable but show lysosomal defects. We show that GARP subunits bind specific sets of Golgi SNAREs within the yeast two-hybrid system. This suggests that the C. elegans GARP complex also facilitates tethering as well as SNARE complex assembly at the Golgi. The GARP and COG tethering complexes may have overlapping functions for retrograde endosome-to-Golgi retrieval, since loss of both complexes leads to a synthetic lethal phenotype.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Golgi Apparatus/metabolism , Lysosomes/ultrastructure , Multiprotein Complexes/metabolism , Vesicular Transport Proteins/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/ultrastructure , Caenorhabditis elegans Proteins/classification , Caenorhabditis elegans Proteins/genetics , Conserved Sequence , Endosomes/genetics , Endosomes/metabolism , Molecular Sequence Data , Multiprotein Complexes/genetics , Phylogeny , SNARE Proteins/metabolism , Transport Vesicles/genetics , Two-Hybrid System Techniques , Vesicular Transport Proteins/classification , Vesicular Transport Proteins/genetics
13.
BMC Genomics ; 11: 671, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-21110867

ABSTRACT

BACKGROUND: Proteins may evolve through the recruitment and modification of discrete domains, and in many cases, protein action can be dissected at the domain level. PDZ domains are found in many important structural and signaling complexes, and are generally thought to interact with their protein partners through a C-terminal consensus sequence. We undertook a comprehensive search for protein partners of all individual PDZ domains in C. elegans to characterize their function and mode of interaction. RESULTS: Coupling high-throughput yeast two-hybrid screens with extensive validation by co-affinity purification, we defined a domain-orientated interactome map. This integrates PDZ domain proteins in numerous cell-signaling pathways and shows that PDZ domain proteins are implicated in an unexpectedly wide range of cellular processes. Importantly, we uncovered a high frequency of non-canonical interactions, not involving the C-terminus of the protein partner, which were directly confirmed in most cases. We completed our study with the generation of a yeast array representing the entire set of PDZ domains from C. elegans and provide a proof-of-principle for its application to the discovery of PDZ domain targets for any protein or peptide of interest. CONCLUSIONS: We provide an extensive domain-centered dataset, together with a clone resource, that will help future functional study of PDZ domains. Through this unbiased approach, we revealed frequent non-canonical interactions between PDZ domains and their protein partners that will require a re-evaluation of this domain's molecular function.[The protein interactions from this publication have been submitted to the IMEx (http://www.imexconsortium.org) consortium through IntAct (PMID: 19850723) and assigned the identifier IM-14654].


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Genome/genetics , PDZ Domains/genetics , Animals , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/classification , Consensus Sequence/genetics , Immunoprecipitation , Protein Binding/genetics , Proteome/chemistry , Proteome/metabolism , Reproducibility of Results , Two-Hybrid System Techniques
14.
Hum Mol Genet ; 19(12): 2347-59, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20233749

ABSTRACT

Mutations of the immunoglobulin superfamily proteins nephrin and Neph1 lead to congenital nephrotic syndrome in humans or mice. Neph proteins are three closely related molecules that are evolutionarily conserved and mediate cell recognition. Their importance for morphogenetic processes including the formation of the kidney filtration barrier in vertebrates and synaptogenesis in Caenorhabditis elegans has recently been uncovered. However, the individual morphogenetic function of mammalian Neph1-3 isoforms remained elusive. We demonstrate now that the Neph/nephrin family proteins can form cell-cell adhesion modules across species. Expression of all three mammalian Neph isoforms partially rescued mutant C. elegans lacking their Neph homolog syg-1 and restored synapse formation, suggesting a functional redundancy between the three isoforms. Strikingly, the rescue of defective synaptic connectivity was prevented by deletion of the highly conserved cytoplasmic PSD95/Dlg/ZO-1-binding motif of SYG-1/Neph proteins, indicating the critical role of this intracellular signaling motif for SYG-1/Neph-dependent morphogenetic events. To determine the significance of Neph isoform redundancy for vertebrate kidney development, we analyzed the expression pattern and the functional role of Neph proteins in zebrafish. In situ hybridizations identified zNeph1 and zNeph2 as glomerular proteins. Morpholino knockdown of either zNeph1 or zNeph2 resulted in loss of slit diaphragms and leakiness of the glomerular filtration barrier. This is the first report utilizing C. elegans to study mammalian Neph/nephrin protein function and to demonstrate a functional overlap of Neph1-3 proteins. Furthermore, we identify Neph2 as a novel critical regulator of glomerular function, indicating that both Neph1 and Neph2 are required for glomerular maintenance and development.


Subject(s)
Immunoglobulins/physiology , Kidney/growth & development , Membrane Proteins/physiology , Neurons/physiology , Amino Acid Sequence , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/classification , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/physiology , Cell Adhesion , HeLa Cells , Humans , Immunoglobulins/classification , Immunoglobulins/genetics , Membrane Proteins/classification , Membrane Proteins/genetics , Mice , Models, Animal , Morphogenesis/genetics , PDZ Domains , Phylogeny
15.
Bioessays ; 32(2): 109-18, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20091748

ABSTRACT

Cold shock domain (CSD)-containing proteins have been found in all three domains of life and function in a variety of processes that are related, for the most part, to post-transcriptional gene regulation. The CSD is an ancient beta-barrel fold that serves to bind nucleic acids. The CSD is structurally and functionally similar to the S1 domain, a fold with otherwise unrelated primary sequence. The flexibility of the CSD/S1 domain for RNA recognition confers an enormous functional versatility to the proteins that contain them. This review summarizes the current knowledge on eukaryotic CSD/S1 domain-containing proteins with a special emphasis on UNR (upstream of N-ras), a member of this family with multiple copies of the CSD.


Subject(s)
Eukaryotic Cells/metabolism , Eukaryotic Initiation Factors/metabolism , Heat-Shock Proteins/metabolism , Animals , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/classification , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Eukaryotic Initiation Factors/chemistry , Eukaryotic Initiation Factors/classification , Eukaryotic Initiation Factors/genetics , Exosomes/metabolism , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/classification , Heat-Shock Proteins/genetics , Humans , Phylogeny , Protein Structure, Secondary , Repressor Proteins/chemistry , Repressor Proteins/classification , Repressor Proteins/genetics , Repressor Proteins/metabolism , Y-Box-Binding Protein 1/chemistry , Y-Box-Binding Protein 1/classification , Y-Box-Binding Protein 1/genetics , Y-Box-Binding Protein 1/metabolism
16.
PLoS Comput Biol ; 5(4): e1000354, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19360093

ABSTRACT

During development, signaling networks control the formation of multicellular patterns. To what extent quantitative fluctuations in these complex networks may affect multicellular phenotype remains unclear. Here, we describe a computational approach to predict and analyze the phenotypic diversity that is accessible to a developmental signaling network. Applying this framework to vulval development in C. elegans, we demonstrate that quantitative changes in the regulatory network can render approximately 500 multicellular phenotypes. This phenotypic capacity is an order-of-magnitude below the theoretical upper limit for this system but yet is large enough to demonstrate that the system is not restricted to a select few outcomes. Using metrics to gauge the robustness of these phenotypes to parameter perturbations, we identify a select subset of novel phenotypes that are the most promising for experimental validation. In addition, our model calculations provide a layout of these phenotypes in network parameter space. Analyzing this landscape of multicellular phenotypes yielded two significant insights. First, we show that experimentally well-established mutant phenotypes may be rendered using non-canonical network perturbations. Second, we show that the predicted multicellular patterns include not only those observed in C. elegans, but also those occurring exclusively in other species of the Caenorhabditis genus. This result demonstrates that quantitative diversification of a common regulatory network is indeed demonstrably sufficient to generate the phenotypic differences observed across three major species within the Caenorhabditis genus. Using our computational framework, we systematically identify the quantitative changes that may have occurred in the regulatory network during the evolution of these species. Our model predictions show that significant phenotypic diversity may be sampled through quantitative variations in the regulatory network without overhauling the core network architecture. Furthermore, by comparing the predicted landscape of phenotypes to multicellular patterns that have been experimentally observed across multiple species, we systematically trace the quantitative regulatory changes that may have occurred during the evolution of the Caenorhabditis genus.


Subject(s)
Caenorhabditis elegans Proteins/classification , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Gene Expression Regulation, Developmental/physiology , Models, Biological , Signal Transduction/physiology , Vulva/physiology , Animals , Caenorhabditis elegans/classification , Computer Simulation , Female , Phenotype , Species Specificity
17.
Mol Cell Biol ; 29(10): 2704-15, 2009 May.
Article in English | MEDLINE | ID: mdl-19273594

ABSTRACT

The transcription factor SKN-1 protects Caenorhabditis elegans from stress and promotes longevity. SKN-1 is regulated by diverse signals that control metabolism, development, and stress responses, but the mechanisms of regulation and signal integration are unknown. We screened the C. elegans genome for regulators of cytoprotective gene expression and identified a new SKN-1 regulatory pathway. SKN-1 protein levels, nuclear accumulation, and activity are repressed by the WD40 repeat protein WDR-23, which interacts with the CUL-4/DDB-1 ubiquitin ligase to presumably target the transcription factor for proteasomal degradation. WDR-23 regulates SKN-1 target genes downstream from p38 mitogen-activated protein kinase, glycogen synthase kinase 3, and insulin-like receptor pathways, suggesting that phosphorylation of SKN-1 may function to modify its interaction with WDR-23 and/or CUL-4/DDB-1. These findings define the mechanism of SKN-1 accumulation in the cell nucleus and provide a new mechanistic framework for understanding how phosphorylation signals are integrated to regulate stress resistance and longevity.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Cell Nucleus/metabolism , Cullin Proteins/metabolism , DNA-Binding Proteins/metabolism , Repetitive Sequences, Amino Acid , Repressor Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/classification , Caenorhabditis elegans Proteins/genetics , Cullin Proteins/genetics , DNA-Binding Proteins/genetics , Forkhead Transcription Factors , Gene Expression Regulation , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Molecular Sequence Data , Phylogeny , Proteasome Endopeptidase Complex/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/classification , Repressor Proteins/genetics , Sequence Alignment , Signal Transduction/physiology , Stress, Physiological , Transcription Factors/genetics , Two-Hybrid System Techniques
18.
J Cell Sci ; 122(Pt 5): 611-24, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19208769

ABSTRACT

Meckel syndrome (MKS) is a ciliopathy characterized by encephalocele, cystic renal disease, liver fibrosis and polydactyly. An identifying feature of MKS1, one of six MKS-associated proteins, is the presence of a B9 domain of unknown function. Using phylogenetic analyses, we show that this domain occurs exclusively within a family of three proteins distributed widely in ciliated organisms. Consistent with a ciliary role, all Caenorhabditis elegans B9-domain-containing proteins, MKS-1 and MKS-1-related proteins 1 and 2 (MKSR-1, MKSR-2), localize to transition zones/basal bodies of sensory cilia. Their subcellular localization is largely co-dependent, pointing to a functional relationship between the proteins. This localization is evolutionarily conserved, because the human orthologues also localize to basal bodies, as well as cilia. As reported for MKS1, disrupting human MKSR1 or MKSR2 causes ciliogenesis defects. By contrast, single, double and triple C. elegans mks/mksr mutants do not display overt defects in ciliary structure, intraflagellar transport or chemosensation. However, we find genetic interactions between all double mks/mksr mutant combinations, manifesting as an increased lifespan phenotype, which is due to abnormal insulin-IGF-I signaling. Our findings therefore demonstrate functional interactions between a novel family of proteins associated with basal bodies or cilia, providing new insights into the molecular etiology of a pleiotropic human disorder.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Cilia/pathology , Proteins/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/classification , Caenorhabditis elegans Proteins/genetics , Cilia/metabolism , Evolution, Molecular , Humans , Molecular Sequence Data , Phenotype , Phylogeny , Proteins/classification , Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Signal Transduction/physiology
19.
PLoS Biol ; 6(9): e239, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18828675

ABSTRACT

PDZ domains are protein-protein interaction modules that recognize specific C-terminal sequences to assemble protein complexes in multicellular organisms. By scanning billions of random peptides, we accurately map binding specificity for approximately half of the over 330 PDZ domains in the human and Caenorhabditis elegans proteomes. The domains recognize features of the last seven ligand positions, and we find 16 distinct specificity classes conserved from worm to human, significantly extending the canonical two-class system based on position -2. Thus, most PDZ domains are not promiscuous, but rather are fine-tuned for specific interactions. Specificity profiling of 91 point mutants of a model PDZ domain reveals that the binding site is highly robust, as all mutants were able to recognize C-terminal peptides. However, many mutations altered specificity for ligand positions both close and far from the mutated position, suggesting that binding specificity can evolve rapidly under mutational pressure. Our specificity map enables the prediction and prioritization of natural protein interactions, which can be used to guide PDZ domain cell biology experiments. Using this approach, we predicted and validated several viral ligands for the PDZ domains of the SCRIB polarity protein. These findings indicate that many viruses produce PDZ ligands that disrupt host protein complexes for their own benefit, and that highly pathogenic strains target PDZ domains involved in cell polarity and growth.


Subject(s)
Caenorhabditis elegans Proteins/analysis , Caenorhabditis elegans Proteins/genetics , PDZ Domains , Proteome/analysis , Amino Acid Sequence , Animals , Binding Sites/genetics , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/classification , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Mutation , Peptides/analysis , Peptides/genetics , Phylogeny , Protein Structure, Secondary , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
20.
Protein Pept Lett ; 15(5): 505-9, 2008.
Article in English | MEDLINE | ID: mdl-18537741

ABSTRACT

Tre-2/Bub2/Cdc16 domain-containing proteins (TBC proteins) participate in wide range cellular processes. With computational approaches, 137 non-redundant TBC proteins from five model organisms were identified and classified into 13 subfamilies base on molecular evolutionary tree. This phylogenetic analysis provides useful functional annotation of newly-identified TBC proteins and guides for further experimentation.


Subject(s)
Amino Acid Motifs , Computational Biology/methods , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/classification , Animals , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/classification , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/classification , Drosophila Proteins/chemistry , Drosophila Proteins/classification , Evolution, Molecular , Humans , Phylogeny , Protein Structure, Tertiary , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/classification , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...