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










Publication year range
1.
Plant Cell Rep ; 43(1): 3, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38117317

ABSTRACT

KEY MESSAGE: Heterologous expression of a nematode-responsive promoter in tomato successfully driven the RNAi constructs to impart root-knot nematode resistance. The root-knot nematode Meloidogyne incognita seriously afflicts the global productivity of tomatoes. Nematode management options are extremely reliant on chemical methods, however, only a handful of nematicides are commercially available. Additionally, nematodes have developed resistance-breaking phenotypes against the commercially available Mi gene-expressing tomatoes. Nematode resistance in crop plants can be enhanced using the bio-safe RNAi technology, in which plants are genetically modified to express nematode gene-specific dsRNA/siRNA molecules. However, the majority of the RNAi crops conferring nematode tolerance have used constitutive promoters, which have many limitations. In the present study, using promoter-GUS fusion, we functionally validated two nematode-inducible root-specific promoters (pAt1g74770 and pAt2g18140, identified from Arabidopsis thaliana) in the Solanum lycopersicum-M. incognita pathosystem. pAt2g18140 was found to be nematode-responsive during 10-21 days post-inoculation (dpi) and became non-responsive during the late infection stage (28 dpi). In contrast, pAt1g74770 remained nematode-responsive for a longer duration (10-28 dpi). Next, a number of transgenic lines were developed that expressed RNAi constructs (independently targeting the M. incognita integrase and splicing factor genes) driven by the pAt1g74770 promoter. M. incognita parasitic success (measured by multiplication factor ratio) in pAt1g74770:integrase and pAt1g74770:splicing factor RNAi lines were significantly reduced by 60.83-74.93% and 69.34-75.31%, respectively, compared to the control. These data were comparable with the RNAi lines having CaMV35S as the promoter. Further, a long-term RNAi effect was evident, because females extracted from transgenic lines were of deformed shape with depleted transcripts of integrase and splicing factor genes. We conclude that pAt1g74770 can be an attractive alternative to drive localized expression of RNAi constructs rather than using a constitutive promoter. The pAt1g74770-driven gene silencing system can be expanded into different plant-nematode interaction models.


Subject(s)
Arabidopsis , Solanum lycopersicum , Tylenchoidea , Female , Animals , RNA Interference , Solanum lycopersicum/genetics , Integrases , RNA Splicing Factors , RNA, Double-Stranded/genetics
2.
Biol Open ; 12(7)2023 07 15.
Article in English | MEDLINE | ID: mdl-37310364

ABSTRACT

Limiting maternal resources necessitates deferring the development of adult-specific structures, notably the reproductive structures, to the postembryonic phase. These structures form postembryonically from blast cells generated during embryogenesis. A close coordination of developmental timing and pattern among the various postembryonic cell lineages is essential to form a functional adult. Here, we show that the C. elegans gene gvd-1 is essential for the development of several structures that form during the late larval stages. In gvd-1 mutant animals, blast cells that normally divide during the late larval stages (L3 and L4) fail to divide. In addition, germ cell proliferation is also severely reduced in these animals. Expression patterns of relevant reporter transgenes revealed a delay in G1/S transition in the vulval precursor cell P6.p and cytokinesis failure in seam cells in gvd-1 larvae. Our analyses of GVD-1::GFP transgenes indicate that GVD-1 is expressed in both soma and germ line, and functions in both. Sequence comparisons revealed that the sequence of gvd-1 is conserved only among nematodes, which does not support a broadly conserved housekeeping function for gvd-1. Instead, our results indicate a crucial role for gvd-1 that is specific to the larval development of nematodes.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Lineage , Germ Cells/metabolism , Cell Proliferation
3.
MicroPubl Biol ; 20222022.
Article in English | MEDLINE | ID: mdl-36338151

ABSTRACT

Mos1 -mediated single-copy integration (MosSCI) in C. elegans relies on the introduction of plasmid constructs into the germ line. Such plasmids form extrachromosomal arrays containing multiple copies of the transgene. Presently, one positive-selection and four negative-selection reporters are used to identify animals that carry the integrated transgene but not the array. Even with four reporters, the negative selection is inefficient. Here, we show that the expression of the toxic protein PEEL-1 from a transgene containing the endogenous peel-1 introns kills all array-carrying animals, which facilitates efficient selection of animals carrying the integrated transgene.

4.
Development ; 147(22)2020 11 27.
Article in English | MEDLINE | ID: mdl-33051256

ABSTRACT

The germline genome is guarded against invading foreign genetic elements by small RNA-dependent gene-silencing pathways. Components of these pathways localize to, or form distinct aggregates in the vicinity of, germ granules. These components and their dynamics in and out of granules are currently being intensively studied. Here, we report the identification of PLP-1, a Caenorhabditiselegans protein related to the human single-stranded nucleic acid-binding protein Pur-alpha, as a component of germ granules in C. elegans We show that PLP-1 is essential for silencing different types of transgenes in the germ line and for suppressing the expression of several endogenous genes controlled by the germline gene-silencing pathways. Our results reveal that PLP-1 functions downstream of small RNA biogenesis during initiation of gene silencing. Based on these results and the earlier findings that Pur-alpha proteins interact with both RNA and protein, we propose that PLP-1 couples certain RNAs with their protein partners in the silencing complex. PLP-1 orthologs localized on RNA granules may similarly contribute to germline gene silencing in other organisms.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , DNA-Binding Proteins/metabolism , Gene Silencing , Germ Cells/metabolism , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , DNA-Binding Proteins/genetics , Germ Cells/cytology , Male
5.
Development ; 145(7)2018 03 29.
Article in English | MEDLINE | ID: mdl-29540500

ABSTRACT

Pairing of homologous chromosomes is essential for genetic recombination during gametogenesis. In many organisms, chromosome ends are attached to cytoplasmic dynein, and dynein-driven chromosomal movements facilitate the pairing process. Factors that promote or control the cytoskeletal tethering of chromosomes are largely unknown. Here, we show that the conserved RNA-binding protein PUF-8 facilitates the tethering and pairing processes in the C. elegans germline by promoting proteasome activity. We have isolated a hypomorphic allele of pas-1, which encodes a proteasome core subunit, and find that the homologous chromosomes fail to pair in the puf-8; pas-1 double mutant due to failure of chromosome tethering. Our results reveal that the puf-8; pas-1 meiotic defects are caused by the loss of proteasome activity. The axis component HTP-3 accumulates prematurely in the double mutant, and reduction of its activity partially suppresses some of the puf-8; pas-1 meiotic defects, suggesting that HTP-3 might be an important target of the proteasome in promoting early meiotic events. In summary, our results reveal a role for the proteasome in chromosome tethering and identify PUF-8 as a regulator of proteasome activity during early meiosis.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Chromosome Pairing/genetics , Meiosis/genetics , Proteasome Endopeptidase Complex/metabolism , RNA-Binding Proteins/metabolism , ATPases Associated with Diverse Cellular Activities/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Cell Cycle Proteins/metabolism , Germ Cells/metabolism , Membrane Proteins/metabolism , Mutagenesis , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
6.
Results Probl Cell Differ ; 59: 175-200, 2017.
Article in English | MEDLINE | ID: mdl-28247049

ABSTRACT

Germline poses unique challenges to gene expression control at the transcriptional level. While the embryonic germline maintains a global hold on new mRNA transcription, the female adult germline produces transcripts that are not translated into proteins until embryogenesis of subsequent generation. As a consequence, translational control plays a central role in governing various germ cell decisions including the formation of primordial germ cells, self-renewal/differentiation decisions in the adult germline, onset of gametogenesis and oocyte maturation. Mechanistically, several common themes such as asymmetric localization of mRNAs, conserved RNA-binding proteins that control translation by 3' UTR binding, translational activation by the cytoplasmic elongation of the polyA tail and the assembly of mRNA-protein complexes called mRNPs have emerged from the studies on Caenorhabditis elegans, Xenopus and Drosophila. How mRNPs assemble, what influences their dynamics, and how a particular 3' UTR-binding protein turns on the translation of certain mRNAs while turning off other mRNAs at the same time and space are key challenges for future work.


Subject(s)
Gene Expression Regulation/genetics , Germ Cells/physiology , Protein Biosynthesis/genetics , Animals , Female , Humans , Male
7.
Front Plant Sci ; 8: 2049, 2017.
Article in English | MEDLINE | ID: mdl-29312363

ABSTRACT

The root-knot nematode (RKN), Meloidogyne incognita, is an obligate, sedentary endoparasite that infects a large number of crops and severely affects productivity. The commonly used nematode control strategies have their own limitations. Of late, RNA interference (RNAi) has become a popular approach for the development of nematode resistance in plants. Transgenic crops capable of expressing dsRNAs, specifically in roots for disrupting the parasitic process, offer an effective and efficient means of producing resistant crops. We identified nematode-responsive and root-specific (NRRS) promoters by using microarray data from the public domain and known conserved cis-elements. A set of 51 NRRS genes was identified which was narrowed down further on the basis of presence of cis-elements combined with minimal expression in the absence of nematode infection. The comparative analysis of promoters from the enriched NRRS set, along with earlier reported nematode-responsive genes, led to the identification of specific cis-elements. The promoters of two candidate genes were used to generate transgenic plants harboring promoter GUS constructs and tested in planta against nematodes. Both promoters showed preferential expression upon nematode infection, exclusively in the root in one and galls in the other. One of these NRRS promoters was used to drive the expression of splicing factor, a nematode-specific gene, for generating host-delivered RNAi-mediated nematode-resistant plants. Transgenic lines expressing dsRNA of splicing factor under the NRRS promoter exhibited upto a 32% reduction in number of galls compared to control plants.

8.
Front Plant Sci ; 8: 2266, 2017.
Article in English | MEDLINE | ID: mdl-29403514

ABSTRACT

Root-knot nematodes have emerged as devastating parasites causing substantial losses to agricultural economy worldwide. Tomato is the most favored host for major species of root-knot nematodes. Control strategies like use of nematicides have proved to be harmful to the environment. Other control methods like development of resistant cultivars and crop rotation have serious limitations. This study deals with the application of host generated RNA interference toward development of resistance against root-knot nematode Meloidogyne incognita in tomato. Two cuticle collagen genes viz. Mi-col-1 and Lemmi-5 involved in the synthesis and maintenance of the cuticle in M. incognita were targeted through host generated RNA interference. Expression of both Mi-col-1 and Lemmi-5 was found to be higher in adult females followed by egg masses and J2s. Tomato var. Pusa Ruby was transformed with the RNAi constructs of these genes to develop transgenic lines expressing the target dsRNAs. 30.80-35.00% reduction in the number of adult females, 50.06-65.73% reduction in the number of egg mass per plant and 76.47-82.59% reduction in the number of eggs per egg mass were observed for the T1 events expressing Mi-col-1 dsRNA. Similarly, 34.14-38.54% reduction in the number of adult females, 62.34-66.71% reduction in number of egg mass per plant and 67.13-79.76% reduction in the number of eggs per egg mass were observed for the T1 generation expressing Lemmi-5 dsRNA. The multiplication factor of M. incognita reduced significantly in both the cases and the structure of adult females isolated from transgenic plants were heavily distorted. This study demonstrates the role of the cuticle collagen genes Mi-col-1 and Lemmi-5 in the structure and development of M. incognita cuticle inside the host and reinforces the potential of host generated RNA interference for management of plant parasitic nematodes (PPNs).

9.
Development ; 143(17): 3097-108, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27510976

ABSTRACT

Membrane-bound receptors, which are crucial for mediating several key developmental signals, are synthesized on endoplasmic reticulum (ER). The functional integrity of ER must therefore be important for the regulation of at least some developmental programs. However, the developmental control of ER function is not well understood. Here, we identify the C. elegans protein FARL-11, an ortholog of the mammalian STRIPAK complex component STRIP1/2 (FAM40A/B), as an ER protein. In the C. elegans embryo, we find that FARL-11 is essential for the cell cycle-dependent morphological changes of ER and for embryonic viability. In the germline, FARL-11 is required for normal ER morphology and for membrane localization of the GLP-1/Notch receptor involved in germline stem cell (GSC) maintenance. Furthermore, we provide evidence that PUF-8, a key translational regulator in the germline, promotes the translation of farl-11 mRNA. These findings reveal that ER form and function in the C. elegans germline are post-transcriptionally regulated and essential for the niche-GSC signaling mediated by GLP-1.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Endoplasmic Reticulum/metabolism , Germ Cells/cytology , Germ Cells/metabolism , Animals , Caenorhabditis elegans Proteins/genetics , Gene Expression Regulation, Developmental/physiology , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/metabolism
10.
G3 (Bethesda) ; 5(8): 1675-84, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-26068572

ABSTRACT

Successful meiotic progression of germ cells is crucial for gametogenesis. Defects in this process affect proper genetic transmission and sometimes lead to tumor formation in the germline. In Caenorhabditis elegans, the RNA-binding protein GLD-1 is essential for the meiotic development of oocytes. However, its role during spermatogenesis has not been understood. Here, we show that GLD-1 functions redundantly with the PUF family protein PUF-8 to ensure proper meiotic development of spermatocytes. When grown at 20°-the standard laboratory temperature for C. elegans growth-primary spermatocytes in both gld-1 and puf-8 single-mutant males and hermaphrodites complete the meiotic divisions normally. By contrast, some of the gld-1; puf-8 double-mutant spermatocytes exit meiosis and form germ cell tumors in both sexes. During larval development, gld-1; puf-8 double-mutant germ cells begin to express the meiotic marker HIM-3, lose P granules, and form the sperm-specific membranous organelle, which are characteristics of developing spermatocytes. However, some of these cells quickly lose HIM-3 and form germ cell tumors that lack membranous organelle but contain P granules. Mutations that block meiotic progression at late pachytene or diakinetic stage fail to arrest the tumorigenesis, suggesting that the gld-1; puf-8 double-mutant spermatocytes exit meiosis prior to the completion of pachytene. Together, results presented here uncover a novel function for gld-1 in the meiotic development of spermatocytes in both hermaphrodites and males.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Meiosis , RNA-Binding Proteins/metabolism , Spermatocytes/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Genes, Reporter , Larva/metabolism , Male , Microscopy, Fluorescence , Mutation , Oocytes/metabolism , RNA Interference , RNA-Binding Proteins/genetics , Spermatocytes/cytology , Spermatogenesis
11.
Development ; 140(8): 1645-54, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23487310

ABSTRACT

Signals that promote germ cell self-renewal by preventing premature meiotic entry are well understood. However, signals that control mitotic proliferation to promote meiotic differentiation have not been well characterized. In Caenorhabditis elegans, GLP-1 Notch signalling promotes the proliferative fate by preventing premature meiotic entry. The germline niche cell, which is the source of the ligand for GLP-1, spatially restricts GLP-1 signalling and thus enables the germ cells that have moved away from the niche to enter meiosis. Here, we show that the suppression of RAS/MAP kinase signalling in the mitotic and meiotic-entry regions is essential for the regulation of the mitosis-meiosis switch by niche signalling. We provide evidence that the conserved PUF family RNA-binding protein PUF-8 and the RAS GAP protein GAP-3 function redundantly to suppress the LET-60 RAS in the mitotic and meiotic entry regions. Germ cells missing both PUF-8 and GAP-3 proliferate in an uncontrolled fashion and fail to undergo meiotic development. MPK-1, the MAP kinase downstream of the LET-60 RAS, is prematurely activated in these cells; downregulation of MPK-1 activation eliminates tumours and restores differentiation. Our results further reveal that PUF-8 negatively regulates LET-60 expression at a post-transcriptional step. LET-60 is misexpressed in the puf-8(-) mutant germlines and PUF-8 physically interacts with the let-60 3' UTR. Furthermore, PUF-8 suppresses let-60 3' UTR-mediated expression in the germ cells that are transitioning from the mitotic to meiotic fate. These results reveal that PUF-8-mediated inhibition of the RAS/MAPK pathway is essential for mitotic-to-meiotic fate transition.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/embryology , Cell Differentiation/physiology , Germ Cells/physiology , Mitogen-Activated Protein Kinase 1/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction/physiology , ras Proteins/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans Proteins/genetics , Electrophoretic Mobility Shift Assay , Immunoprecipitation , Microscopy, Fluorescence , Mutagenesis , RNA-Binding Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
12.
Development ; 140(6): 1312-20, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23444359

ABSTRACT

PUF family proteins are well-conserved regulators of cell proliferation in different developmental processes. They regulate target mRNAs by promoting degradation or by influencing translation through interaction with the translation initiation machinery. Here we show that Caenorhabditis elegans PUF-8 functions redundantly with the nuclear protein TCER-1 in the post-transcriptional maintenance of at least six germline mRNAs. The levels of spliced mRNAs in the puf-8(-) tcer-1(-) double mutant are only 10-30% of the wild type, whereas the unspliced forms increase by ∼2- to 3-fold compared with the wild type. These two proteins colocalise at the inner nuclear periphery, and their absence leads to reduced germ cell proliferation and to sterility. A yeast two-hybrid screen of 31 components of the nuclear pore complex and mRNA processing machineries identified seven proteins involved in mRNA export as potential partners of PUF-8. One of these, the nuclear cap-binding protein NCBP-2, colocalises with PUF-8 in the nucleus. A 50 amino acid N-terminal domain of PUF-8 is essential for interaction with NCBP-2 and for PUF-8 to function redundantly with TCER-1. These results reveal two important unexpected aspects of PUF proteins: that, in addition to the C-terminal PUF domain, the N-terminal domain is crucial for PUF function, and that PUF proteins have a novel role in mRNA maintenance. We propose that PUF proteins, in addition to their known cytoplasmic roles, participate in nuclear processing and/or export of mRNAs.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans , Germ Cells/metabolism , Peptide Elongation Factors/physiology , RNA, Messenger/genetics , RNA-Binding Proteins/physiology , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Gene Expression Regulation, Developmental , Germ Cells/growth & development , Peptide Elongation Factors/genetics , Peptide Elongation Factors/metabolism , Protein Binding , RNA Processing, Post-Transcriptional/genetics , RNA Transport/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Tissue Distribution , Two-Hybrid System Techniques
13.
Mol Biochem Parasitol ; 186(1): 51-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23043990

ABSTRACT

Many of the currently available nematicides used in nematode control are hazardous to the user, environment and beneficial non-target organisms. Therefore the need to develop alternative methods for nematode control such as the development of nematode-resistant crops through RNA-mediated interference (RNAi) holds great promise. The Caenorhabditis elegans genes unc-87 and pat-10 are essential components of the body wall muscle and are thus required for nematode movement. The Pratylenchus coffeae orthologs of these two genes, namely Pc-pat-10 and Pc-unc-87 were cloned and used to test RNAi in this migratory nematode. RNAi was performed by soaking P. coffeae in a solution containing dsRNA of either Pc-unc-87 or Pc-pat-10. The levels of both Pc-unc-87 and Pc-pat-10 mRNAs were significantly reduced in a sequence-specific manner in nematodes soaked for 24h. Nematodes incubated in Pc-pat-10 dsRNA appeared straight and rigid while Pc-unc-87 resulted in nematodes that were coiled, in contrast to the regular sinusoidal movement of the control nematodes. While 88.4 ± 3.9% of the control nematodes successfully migrated to the bottom of the sand column in 12h, only 6 ± 1.3% and 7 ± 2.3%, respectively, of the Pc-pat-10 (RNAi) and Pc-unc-87 (RNAi) nematodes successfully migrated to the bottom. However a recovery in movement as well as transcript level was observed in both treatments when the nematodes were incubated in distilled water for 24h following the dsRNA soaking. The recovery rate was slower in Pc-unc-87 when compared to Pc-pat-10. In summary, this study demonstrates the existence of the RNAi phenomenon in P. coffeae and shows that the function of unc-87 and pat-10 genes has been evolutionarily conserved among free-living and plant parasitic nematodes.


Subject(s)
Helminth Proteins/genetics , Locomotion , RNA Interference , Secernentea Infections/pathology , Tylenchoidea/physiology , Animals , Helminth Proteins/metabolism , Tylenchoidea/genetics
14.
Dev Biol ; 360(1): 195-207, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21968099

ABSTRACT

RNA-binding proteins of the PUF family are well conserved post-transcriptional regulators that control a variety of developmental processes. The C. elegans protein PUF-8 is essential for several aspects of germ cell development including the maintenance of germline stem cells (GSCs). To explore the molecular mechanisms underlying its function, we have identified 160 germline-expressed mRNAs as potential targets of PUF-8. We generated GFP::H2B-3' UTR fusions for 17 mRNAs to assay their post-transcriptional regulation in germ cells. Twelve transgenes were not expressed in the mitotic germ cells, and depletion of PUF-8 led to misexpression of six of them in these cells. In contrast, the expression of 3' UTR fusion of hip-1, which encodes the HSP-70 interacting protein, was dependent on PUF-8. These results indicate that PUF-8 may regulate the expression of its targets both negatively as well as positively. We investigated the PUF-8-mediated post-transcriptional control of one mRNA, namely pal-1, which encodes a homeodomain transcription factor responsible for muscle development. Our results show that PUF-8 binds in vitro to specific sequences within pal-1 3' UTR that are critical for post-transcriptional suppression in GSCs. Removal of PUF-8 resulted in PAL-1 misexpression, and PAL-1-dependent misexpression of the myogenic promoter HLH-1 in germ cells. We propose that PUF-8 protects GSCs from the influence of somatic differentiation factors such as PAL-1, which are produced in the maternal germline but meant for embryogenesis.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism , Embryonic Stem Cells/metabolism , Germ Cells/metabolism , Homeodomain Proteins/metabolism , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , 3' Untranslated Regions , Animals , Animals, Genetically Modified , Base Sequence , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Embryonic Stem Cells/cytology , Female , Gene Expression Regulation, Developmental , Genes, Helminth , Germ Cells/cytology , Homeodomain Proteins/genetics , Male , Mitosis , Muscle Proteins , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Nuclear Proteins , RNA Interference , RNA Processing, Post-Transcriptional , RNA, Helminth/genetics , RNA, Helminth/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Trans-Activators/genetics , Transcription Factors
15.
Dev Biol ; 326(2): 295-304, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19100255

ABSTRACT

Maintenance of mitotically cycling germline stem cells (GSCs) is vital for continuous production of gametes. In worms and insects, signaling from surrounding somatic cells play an essential role in the maintenance of GSCs by preventing premature differentiation. In addition, germ cell proteins such as the Drosophila Pumilio and Caenorhabditis elegans FBF, both members of the PUF family translational regulators, contribute to GSC maintenance. FBF functions by suppressing GLD-1, which promotes meiotic entry. However, factors that directly promote GSC proliferation, rather than prevent differentiation, are not known. Here we show that PUF-8, another C. elegans member of the PUF family and MEX-3, a KH domain translational regulator, function redundantly to promote GSC mitosis. We find that PUF-8 protein is highly enriched in mitotic germ cells, which is similar to the expression pattern of MEX-3 described earlier. The puf-8(-) mex-3(-) double mutant gonads contain far fewer germ cells than both single mutants and wild-type. While these cells lack mitotic, meiotic and sperm markers, they retain the germ cell-specific P granules, and are capable of gametogenesis if GLP-1, which normally blocks meiotic entry, is removed. Significantly, we find that at least one of these two proteins is essential for germ cell proliferation even in meiotic entry-defective mutants, which otherwise produce germ cell tumors. We conclude PUF-8 and MEX-3 contribute to GSC maintenance by promoting mitotic proliferation rather than by blocking meiotic entry.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans , Germ Cells/physiology , Mitosis/physiology , RNA-Binding Proteins/metabolism , Stem Cells/physiology , Animals , Animals, Genetically Modified , Caenorhabditis elegans/cytology , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Cell Differentiation , Cell Proliferation , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Female , Germ Cells/cytology , Male , Meiosis/physiology , RNA-Binding Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Stem Cells/cytology
16.
Development ; 135(10): 1803-12, 2008 May.
Article in English | MEDLINE | ID: mdl-18417623

ABSTRACT

Although germ cell formation has been relatively well understood in worms and insects, how germ cell-specific developmental programs are initiated is not clear. In Caenorhabditis elegans, translational activation of maternal nos-2 mRNA is the earliest known molecular event specific to the germline founder cell P(4). Cis-elements in nos-2 3'UTR have been shown to mediate translational control; however, the trans-acting proteins are not known. Here, we provide evidence that four maternal RNA-binding proteins, OMA-1, OMA-2, MEX-3 and SPN-4, bind nos-2 3'UTR to suppress its translation, and POS-1, another maternal RNA-binding protein, relieves this suppression in P(4). The POS-1: SPN-4 ratio in P(4) increases significantly over its precursor, P(3); and POS-1 competes with SPN-4 for binding to nos-2 RNA in vitro. We propose temporal changes in the relative concentrations of POS-1 and SPN-4, through their effect on the translational status of maternal mRNAs such as nos-2, initiate germ cell-specific developmental programs in C. elegans.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Germ Cells/physiology , 3' Untranslated Regions , Animals , Animals, Genetically Modified , Base Sequence , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/biosynthesis , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Carrier Proteins/metabolism , Carrier Proteins/physiology , Cell Cycle Proteins/metabolism , Gene Expression Regulation, Developmental , Germ Cells/metabolism , Molecular Sequence Data , Oocytes/growth & development , Oocytes/physiology , RNA-Binding Proteins/metabolism
18.
Dev Biol ; 292(1): 244-52, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16499902

ABSTRACT

Members of the nanos gene family are evolutionarily conserved regulators of germ cell development. In several organisms, Nanos protein expression is restricted to the primordial germ cells (PGCs) during early embryogenesis. Here, we investigate the regulation of the Caenorhabditis elegans nanos homolog nos-2. We find that the nos-2 RNA is translationally repressed. In the adult germline, translation of the nos-2 RNA is inhibited in growing oocytes, and this inhibition depends on a short stem loop in the nos-2 3'UTR. In embryos, nos-2 translation is repressed in early blastomeres, and this inhibition depends on a second region in the nos-2 3'UTR. nos-2 RNA is also degraded in somatic blastomeres by a process that is independent of translational repression and requires the CCCH finger proteins MEX-5 and MEX-6. Finally, the germ plasm component POS-1 activates nos-2 translation in the PGCs. A combination of translational repression, RNA degradation, and activation by germ plasm has also been implicated in the regulation of nanos homologs in Drosophila and zebrafish, suggesting the existence of conserved mechanisms to restrict Nanos expression to the germline.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/embryology , Gene Expression Regulation, Developmental/physiology , Protein Biosynthesis/genetics , 3' Untranslated Regions/physiology , Animals , Base Sequence , Blastomeres/metabolism , Caenorhabditis elegans/chemistry , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/biosynthesis , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/physiology , Carrier Proteins/physiology , Cell Lineage/physiology , Germ Cells/metabolism , Molecular Sequence Data , RNA, Messenger/metabolism , RNA-Binding Proteins , Structural Homology, Protein , Transgenes
19.
Curr Biol ; 13(2): 134-9, 2003 Jan 21.
Article in English | MEDLINE | ID: mdl-12546787

ABSTRACT

PUF proteins are a conserved family of RNA binding proteins that regulate RNA stability and translation by binding to specific sequences in 3'-untranslated regions. Drosophila PUMILIO and C. elegans FBF are essential for self-renewal of germline stem cells, suggesting that a common function of PUF proteins may be to sustain mitotic proliferation of stem cells. Here, we show that PUF-8, the C. elegans PUF most related to PUMILIO, performs a different function in germ cells that have begun meiosis: in primary spermatocytes, puf-8 is required to maintain meiosis and prevent the return to mitosis. Primary spermatocytes lacking PUF-8 complete meiotic prophase but do not undergo normal meiotic divisions. Instead, they dedifferentiate back into mitotically cycling germ cells and form rapidly growing tumors. These findings reveal an unexpected ability for germ cells that have completed meiotic prophase to return to the mitotic cycle, and they support the view that PUF proteins regulate multiple transitions during germline development.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Spermatocytes/cytology , Spermatocytes/metabolism , Animals , Caenorhabditis elegans/growth & development , Cell Differentiation , Genes, Helminth , Germinoma/genetics , Germinoma/pathology , Male , Meiosis/genetics , Mitosis/genetics , Spermatogenesis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...