Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Nucleic Acids Res ; 52(9): 4985-5001, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38471816

ABSTRACT

Many microRNA (miRNA)-guided Argonaute proteins can cleave RNA ('slicing'), even though miRNA-mediated target repression is generally cleavage-independent. Here we use Caenorhabditis elegans to examine the role of catalytic residues of miRNA Argonautes in organismal development. In contrast to previous work, mutations in presumed catalytic residues did not interfere with development when introduced by CRISPR. We find that unwinding and decay of miRNA star strands is weakly defective in the catalytic residue mutants, with the largest effect observed in embryos. Argonaute-Like Gene 2 (ALG-2) is more dependent on catalytic residues for unwinding than ALG-1. The miRNAs that displayed the greatest (albeit minor) dependence on catalytic residues for unwinding tend to form stable duplexes with their star strand, and in some cases, lowering duplex stability alleviates dependence on catalytic residues. While a few miRNA guide strands are reduced in the mutant background, the basis of this is unclear since changes were not dependent on EBAX-1, an effector of Target-Directed miRNA Degradation (TDMD). Overall, this work defines a role for the catalytic residues of miRNA Argonautes in star strand decay; future work should examine whether this role contributes to the selection pressure to conserve catalytic activity of miRNA Argonautes across the metazoan phylogeny.


Subject(s)
Argonaute Proteins , Caenorhabditis elegans Proteins , Caenorhabditis elegans , MicroRNAs , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/chemistry , Argonaute Proteins/metabolism , Argonaute Proteins/genetics , Argonaute Proteins/chemistry , RNA Stability/genetics , Mutation , Catalytic Domain/genetics , CRISPR-Cas Systems , RNA-Binding Proteins
2.
bioRxiv ; 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-36711716

ABSTRACT

Many Argonaute proteins can cleave RNA ("slicing") as part of the microRNA-induced silencing complex (miRISC), even though miRNA-mediated target repression is generally independent of target cleavage. Here we use genome editing in C. elegans to examine the role of miRNA-guided slicing in organismal development. In contrast to previous work, slicing-inactivating mutations did not interfere with normal development when introduced by CRISPR. We find that unwinding and decay of miRNA star strands is weakly defective in the absence of slicing, with the largest effect observed in embryos. Argonaute-Like Gene 2 (ALG-2) is more dependent on slicing for unwinding than ALG-1. The miRNAs that displayed the greatest (albeit minor) dependence on slicing for unwinding tend to form stable duplexes with their star strand, and in some cases, lowering duplex stability alleviates dependence on slicing. Gene expression changes were consistent with negligible to moderate loss of function for miRNA guides whose star strand was upregulated, suggesting a reduced proportion of mature miRISC in slicing mutants. While a few miRNA guide strands are reduced in the mutant background, the basis of this is unclear since changes were not dependent on EBAX-1, a factor in the Target-Directed miRNA Degradation (TDMD) pathway. Overall, this work defines a role for miRNA Argonaute slicing in star strand decay; future work should examine whether this role could have contributed to the selection pressure to conserve catalytic activity of miRNA Argonautes across the metazoan phylogeny.

3.
Mol Biol Cell ; 34(9): br15, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37342879

ABSTRACT

Centrosomes are essential parts of diverse cellular processes, and precise regulation of the levels of their constituent proteins is critical for their function. One such protein is Pericentrin (PCNT) in humans and Pericentrin-like protein (PLP) in Drosophila. Increased PCNT expression and its protein accumulation are linked to clinical conditions including cancer, mental disorders, and ciliopathies. However, the mechanisms by which PCNT levels are regulated remain underexplored. Our previous study demonstrated that PLP levels are sharply down-regulated during early spermatogenesis and this regulation is essential to spatially position PLP on the proximal end of centrioles. We hypothesized that the sharp drop in PLP protein was a result of rapid protein degradation during the male germ line premeiotic G2 phase. Here, we show that PLP is subject to ubiquitin-mediated degradation and identify multiple proteins that promote the reduction of PLP levels in spermatocytes, including the UBR box containing E3 ligase Poe (UBR4), which we show binds to PLP. Although protein sequences governing posttranslational regulation of PLP are not restricted to a single region of the protein, we identify a region that is required for Poe-mediated degradation. Experimentally stabilizing PLP, via internal PLP deletions or loss of Poe, leads to PLP accumulation in spermatocytes, its mispositioning along centrioles, and defects in centriole docking in spermatids.


Subject(s)
Centrioles , Ubiquitin-Protein Ligases , Male , Humans , Ubiquitin-Protein Ligases/metabolism , Centrioles/metabolism , Centrosome/metabolism , Antigens/metabolism
4.
Cell Rep ; 40(6): 111154, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35947946

ABSTRACT

MicroRNA (miRNA) abundance is tightly controlled by regulation of biogenesis and decay. Here, we show that the mir-35 miRNA family undergoes selective decay at the transition from embryonic to larval development in C. elegans. The seed sequence of the miRNA is necessary and largely sufficient for this regulation. Sequences outside the seed (3' end) regulate mir-35 abundance in the embryo but are not necessary for sharp decay at the transition to larval development. Enzymatic modifications of the miRNA 3' end are neither prevalent nor correlated with changes in decay, suggesting that miRNA 3' end display is not a core feature of this mechanism and further supporting a seed-driven decay model. Our findings demonstrate that seed-sequence-specific decay can selectively and coherently regulate all redundant members of a miRNA seed family, a class of mechanism that has great biological and therapeutic potential for dynamic regulation of a miRNA family's target repertoire.


Subject(s)
Caenorhabditis elegans Proteins , MicroRNAs , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , MicroRNAs/genetics
6.
Nucleic Acids Res ; 49(19): 11167-11180, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34586415

ABSTRACT

microRNAs are frequently modified by addition of untemplated nucleotides to the 3' end, but the role of this tailing is often unclear. Here we characterize the prevalence and functional consequences of microRNA tailing in vivo, using Caenorhabditis elegans. MicroRNA tailing in C. elegans consists mostly of mono-uridylation of mature microRNA species, with rarer mono-adenylation which is likely added to microRNA precursors. Through a targeted RNAi screen, we discover that the TUT4/TUT7 gene family member CID-1/CDE-1/PUP-1 is required for uridylation, whereas the GLD2 gene family member F31C3.2-here named GLD-2-related 2 (GLDR-2)-is required for adenylation. Thus, the TUT4/TUT7 and GLD2 gene families have broadly conserved roles in miRNA modification. We specifically examine the role of tailing in microRNA turnover. We determine half-lives of microRNAs after acute inactivation of microRNA biogenesis, revealing that half-lives are generally long (median = 20.7 h), as observed in other systems. Although we observe that the proportion of tailed species increases over time after biogenesis, disrupting tailing does not alter microRNA decay. Thus, tailing is not a global regulator of decay in C. elegans. Nonetheless, by identifying the responsible enzymes, this study lays the groundwork to explore whether tailing plays more specialized context- or miRNA-specific regulatory roles.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Cell Cycle Proteins/genetics , Genome, Helminth , MicroRNAs/genetics , RNA, Helminth/genetics , Uridine Monophosphate/metabolism , Adenosine Monophosphate/metabolism , Animals , Caenorhabditis elegans/classification , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Cell Cycle Proteins/metabolism , Chickens/classification , Chickens/genetics , Chickens/metabolism , Conserved Sequence , Gene Expression Regulation , Half-Life , Humans , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/classification , MicroRNAs/metabolism , Phylogeny , RNA Interference , RNA Stability , RNA, Helminth/classification , RNA, Helminth/metabolism , Species Specificity , Zebrafish/classification , Zebrafish/genetics , Zebrafish/metabolism
7.
Genes Dev ; 34(17-18): 1227-1238, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32820039

ABSTRACT

Identifying miRNA target genes is difficult, and delineating which targets are the most biologically important is even more difficult. We devised a novel strategy to test the phenotypic impact of individual microRNA-target interactions by disrupting each predicted miRNA-binding site by CRISPR-Cas9 genome editing in C. elegans We developed a multiplexed negative selection screening approach in which edited loci are deep sequenced, and candidate sites are prioritized based on apparent selection pressure against mutations that disrupt miRNA binding. Importantly, our screen was conducted in vivo on mutant animals, allowing us to interrogate organism-level phenotypes. We used this approach to screen for phenotypic targets of the essential mir-35-42 family. By generating 1130 novel 3'UTR alleles across all predicted targets, we identified egl-1 as a phenotypic target whose derepression partially phenocopies the mir-35-42 mutant phenotype by inducing embryonic lethality and low fecundity. These phenotypes can be rescued by compensatory CRISPR mutations that retarget mir-35 to the mutant egl-1 3'UTR. This study demonstrates that the application of in vivo whole organismal CRISPR screening has great potential to accelerate the discovery of phenotypic negative regulatory elements in the noncoding genome.


Subject(s)
Caenorhabditis elegans/genetics , MicroRNAs/metabolism , 3' Untranslated Regions/genetics , Alleles , Animals , Binding Sites/genetics , CRISPR-Cas Systems , Gene Editing , Genetic Testing , MicroRNAs/genetics , Mutation , Phenotype
9.
FEBS J ; 287(14): 2914-2922, 2020 07.
Article in English | MEDLINE | ID: mdl-31975506

ABSTRACT

Identifying microRNA (miRNA) target genes remains a major challenge in understanding the roles miRNAs play in gene regulation. Furthermore, understanding which miRNA-target interactions are the most biologically important is even more difficult. We present CRISPR-based strategies to identify essential miRNA binding sites. First, CRISPR knockout screens can easily be adapted to identify genes whose inactivation suppresses miRNA mutant phenotypes. Second, a custom approach to target individual miRNA binding sites via CRISPR can identify sites whose mutation recapitulates miRNA mutant phenotypes. We emphasize that the latter approach requires a readout of mutational profile (rather than single guide RNA abundance) when applied in a negative selection setting. Overall, the advent of CRISPR technology alongside improving empirical means of miRNA target identification will accelerate our dissection of miRNA gene regulatory networks.


Subject(s)
CRISPR-Cas Systems , Computational Biology/methods , Gene Expression Regulation , MicroRNAs/genetics , RNA, Messenger/metabolism , Humans , Phenotype , RNA, Messenger/genetics
10.
G3 (Bethesda) ; 9(3): 901-909, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30679246

ABSTRACT

The germline sex determination pathway in C. elegans determines whether germ cells develop as oocytes or sperm, with no previously known effect on viability. The mir-35 family of microRNAs are expressed in the C. elegans germline and embryo and are essential for both viability and normal hermaphroditic sex determination, preventing aberrant male gene expression in XX hermaphrodite embryos. Here we show that combining feminizing mutations with partial loss of function of the mir-35 family results in enhanced penetrance embryonic lethality that preferentially kills XO animals. This lethal phenotype is due to altered signaling through the germline sex determination pathway, and maternal germline feminization is sufficient to induce enhanced lethality. These findings reveal a surprising pleiotropy of sperm-fate promoting pathways on organismal viability. Overall, our results demonstrate an unexpectedly strong link between sex determination and embryonic viability, and suggest that in wild type animals, mir-35 family members buffer against misregulation of pathways outside the sex determination program, allowing for clean sex reversal rather than deleterious effects of perturbing sex determination genes.


Subject(s)
Caenorhabditis elegans/genetics , Gene Expression Regulation, Developmental , Germ Cells/metabolism , MicroRNAs/metabolism , Sex Determination Processes , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/physiology , Hermaphroditic Organisms , Male , MicroRNAs/physiology
11.
Elife ; 72018 12 21.
Article in English | MEDLINE | ID: mdl-30575518

ABSTRACT

Proper regulation of germline gene expression is essential for fertility and maintaining species integrity. In the C. elegans germline, a diverse repertoire of regulatory pathways promote the expression of endogenous germline genes and limit the expression of deleterious transcripts to maintain genome homeostasis. Here we show that the conserved TRIM-NHL protein, NHL-2, plays an essential role in the C. elegans germline, modulating germline chromatin and meiotic chromosome organization. We uncover a role for NHL-2 as a co-factor in both positively (CSR-1) and negatively (HRDE-1) acting germline 22G-small RNA pathways and the somatic nuclear RNAi pathway. Furthermore, we demonstrate that NHL-2 is a bona fide RNA binding protein and, along with RNA-seq data point to a small RNA independent role for NHL-2 in regulating transcripts at the level of RNA stability. Collectively, our data implicate NHL-2 as an essential hub of gene regulatory activity in both the germline and soma.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Carrier Proteins/metabolism , Germ Cells/metabolism , RNA Interference , Animals , Chromatin/metabolism , Gene Regulatory Networks
12.
RNA Biol ; 15(2): 165-169, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29120257

ABSTRACT

The window of embryonic development after fertilization but prior to the beginning of transcription from the zygotic genome is a period that relies heavily on post-transcriptional regulation of gene expression. MicroRNAs constitute one of the predominant mechanisms of post-transcriptional gene regulation, yet their biological function and molecular mechanism of action during this developmental window is poorly understood. Our recent findings demonstrate that the maternal contribution of mir-35 family members contributes to zygotic developmental decisions (sex determination) in C. elegans embryogenesis. Here, I discuss these finding in the context of data from C. elegans and other model organisms regarding the regulation of maternal microRNA activity in early animal embryogenesis.


Subject(s)
Caenorhabditis elegans/embryology , Maternal Inheritance , MicroRNAs/genetics , Animals , Caenorhabditis elegans/genetics , Embryonic Development , Female , Gene Expression Regulation, Developmental , Sex Determination Processes
13.
Genes Dev ; 31(4): 422-437, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28279983

ABSTRACT

Gene expression in early animal embryogenesis is in large part controlled post-transcriptionally. Maternally contributed microRNAs may therefore play important roles in early development. We elucidated a major biological role of the nematode mir-35 family of maternally contributed essential microRNAs. We show that this microRNA family regulates the sex determination pathway at multiple levels, acting both upstream of and downstream from her-1 to prevent aberrantly activated male developmental programs in hermaphrodite embryos. Both of the predicted target genes that act downstream from the mir-35 family in this process, suppressor-26 (sup-26) and NHL (NCL-1, HT2A, and LIN-41 repeat) domain-containing-2 (nhl-2), encode RNA-binding proteins, thus delineating a previously unknown post-transcriptional regulatory subnetwork within the well-studied sex determination pathway of Caenorhabditis elegans Repression of nhl-2 by the mir-35 family is required for not only proper sex determination but also viability, showing that a single microRNA target site can be essential. Since sex determination in C. elegans requires zygotic gene expression to read the sex chromosome karyotype, early embryos must remain gender-naïve; our findings show that the mir-35 family microRNAs act in the early embryo to function as a developmental timer that preserves naïveté and prevents premature deleterious developmental decisions.


Subject(s)
Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Embryonic Development/genetics , Gene Expression Regulation, Developmental/genetics , MicroRNAs/metabolism , Sex Determination Processes/genetics , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Female , Male , MicroRNAs/genetics , Mutation , RNA-Binding Proteins/metabolism
14.
Genes Dev ; 31(2): 209-222, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28167500

ABSTRACT

Programmed cell death occurs in a highly reproducible manner during Caenorhabditis elegans development. We demonstrate that, during embryogenesis, miR-35 and miR-58 bantam family microRNAs (miRNAs) cooperate to prevent the precocious death of mothers of cells programmed to die by repressing the gene egl-1, which encodes a proapoptotic BH3-only protein. In addition, we present evidence that repression of egl-1 is dependent on binding sites for miR-35 and miR-58 family miRNAs within the egl-1 3' untranslated region (UTR), which affect both mRNA copy number and translation. Furthermore, using single-molecule RNA fluorescent in situ hybridization (smRNA FISH), we show that egl-1 is transcribed in the mother of a cell programmed to die and that miR-35 and miR-58 family miRNAs prevent this mother from dying by keeping the copy number of egl-1 mRNA below a critical threshold. Finally, miR-35 and miR-58 family miRNAs can also dampen the transcriptional boost of egl-1 that occurs specifically in a daughter cell that is programmed to die. We propose that miRNAs compensate for lineage-specific differences in egl-1 transcriptional activation, thus ensuring that EGL-1 activity reaches the threshold necessary to trigger death only in daughter cells that are programmed to die.


Subject(s)
Apoptosis/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/genetics , MicroRNAs/metabolism , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Lineage , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , MicroRNAs/genetics , Mutation , Phenotype
16.
PLoS Genet ; 12(12): e1006484, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27935964

ABSTRACT

MicroRNAs and Argonaute form the microRNA induced silencing complex or miRISC that recruits GW182, causing mRNA degradation and/or translational repression. Despite the clear conservation and molecular significance, it is unknown if miRISC-GW182 interaction is essential for gene silencing during animal development. Using Caenorhabditis elegans to explore this question, we examined the relationship and effect on gene silencing between the GW182 orthologs, AIN-1 and AIN-2, and the microRNA-specific Argonaute, ALG-1. Homology modeling based on human Argonaute structures indicated that ALG-1 possesses conserved Tryptophan-binding Pockets required for GW182 binding. We show in vitro and in vivo that their mutations severely altered the association with AIN-1 and AIN-2. ALG-1 tryptophan-binding pockets mutant animals retained microRNA-binding and processing ability, but were deficient in reporter silencing activity. Interestingly, the ALG-1 tryptophan-binding pockets mutant phenocopied the loss of alg-1 in worms during larval stages, yet was sufficient to rescue embryonic lethality, indicating the dispensability of AINs association with the miRISC at this developmental stage. The dispensability of AINs in miRNA regulation is further demonstrated by the capacity of ALG-1 tryptophan-binding pockets mutant to regulate a target of the embryonic mir-35 microRNA family. Thus, our results demonstrate that the microRNA pathway can act independently of GW182 proteins during C. elegans embryogenesis.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Carrier Proteins/genetics , Embryonic Development/genetics , MicroRNAs/genetics , RNA-Binding Proteins/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/chemistry , Gene Expression Regulation, Developmental , Humans , MicroRNAs/biosynthesis , Mutation , RNA Interference , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/chemistry , RNA-Induced Silencing Complex/genetics
17.
Nat Med ; 20(10): 1138-46, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25216638

ABSTRACT

In solid tumors, resistance to therapy inevitably develops upon treatment with cytotoxic drugs or molecularly targeted therapies. Here, we describe a system that enables pooled shRNA screening directly in mouse hepatocellular carcinomas (HCC) in vivo to identify genes likely to be involved in therapy resistance. Using a focused shRNA library targeting genes located within focal genomic amplifications of human HCC, we screened for genes whose inhibition increased the therapeutic efficacy of the multikinase inhibitor sorafenib. Both shRNA-mediated and pharmacological silencing of Mapk14 (p38α) were found to sensitize mouse HCC to sorafenib therapy and prolong survival by abrogating Mapk14-dependent activation of Mek-Erk and Atf2 signaling. Elevated Mapk14-Atf2 signaling predicted poor response to sorafenib therapy in human HCC, and sorafenib resistance of p-Mapk14-expressing HCC cells could be reverted by silencing Mapk14. Our results suggest that a combination of sorafenib and Mapk14 blockade is a promising approach to overcoming therapy resistance of human HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/genetics , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/genetics , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Activating Transcription Factor 2/metabolism , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Drug Resistance, Neoplasm/genetics , Female , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms, Experimental/metabolism , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 14/metabolism , Niacinamide/pharmacology , Protein Kinase Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction , Sorafenib , Xenograft Model Antitumor Assays
18.
G3 (Bethesda) ; 4(9): 1747-54, 2014 Jul 21.
Article in English | MEDLINE | ID: mdl-25053708

ABSTRACT

MicroRNAs guide many aspects of development in all metazoan species. Frequently, microRNAs are expressed during a specific developmental stage to perform a temporally defined function. The C. elegans mir-35-42 microRNAs are expressed abundantly in oocytes and early embryos and are essential for embryonic development. Here, we show that these embryonic microRNAs surprisingly also function to control the number of progeny produced by adult hermaphrodites. Using a temperature-sensitive mir-35-42 family mutant (a deletion of the mir-35-41 cluster), we demonstrate three distinct defects in hermaphrodite fecundity. At permissive temperatures, a mild sperm defect partially reduces hermaphrodite fecundity. At restrictive temperatures, somatic gonad dysfunction combined with a severe sperm defect sharply reduces fecundity. Multiple lines of evidence, including a late embryonic temperature-sensitive period, support a role for mir-35-41 early during development to promote subsequent sperm production in later larval stages. We further show that the predicted mir-35 family target sup-26 (suppressor-26) acts downstream of mir-35-41 in this process, suggesting that sup-26 de-repression in mir-35-41 deletion mutants may contribute to temperature-sensitive loss of fecundity. In addition, these microRNAs play a role in male fertility, promoting proper morphogenesis of male-specific mating structures. Overall, our results demonstrate that robust activity of the mir-35-42 family microRNAs not only is essential for embryonic development across a range of temperatures but also enables the worm to subsequently develop full reproductive capacity.


Subject(s)
Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , MicroRNAs/physiology , Animals , Embryonic Development , Female , Fertility/genetics , Hermaphroditic Organisms , Male , Morphogenesis , Spermatogenesis/genetics , Tail/growth & development , Temperature
19.
Nat Protoc ; 7(2): 374-93, 2012 Feb 02.
Article in English | MEDLINE | ID: mdl-22301776

ABSTRACT

RNA interference (RNAi) is an extremely effective tool for studying gene function in almost all metazoan and eukaryotic model systems. RNAi in mice, through the expression of short hairpin RNAs (shRNAs), offers something not easily achieved with traditional genetic approaches-inducible and reversible gene silencing. However, technical variability associated with the production of shRNA transgenic strains has so far limited their widespread use. Here we describe a pipeline for the generation of miR30-based shRNA transgenic mice that enables efficient and consistent targeting of doxycycline-regulated, fluorescence-linked shRNAs to the Col1a1 locus. Notably, the protocol details crucial steps in the design and testing of miR30-based shRNAs to maximize the potential for developing effective transgenic strains. In all, this 14-week procedure provides a fast and cost-effective way for any laboratory to investigate gene function in vivo in the mouse.


Subject(s)
Genetic Engineering/methods , Mice, Transgenic , RNA Interference , RNA, Small Interfering/genetics , Animals , Cloning, Molecular , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Genotyping Techniques , Mice , MicroRNAs/genetics
20.
Proc Natl Acad Sci U S A ; 108(17): 7113-8, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21482754

ABSTRACT

RNAi has revolutionized loss-of-function genetics by enabling sequence-specific suppression of virtually any gene. Furthermore, tetracycline response elements (TRE) can drive expression of short hairpin RNAs (shRNAs) for inducible and reversible target gene suppression. Here, we demonstrate the feasibility of transgenic inducible RNAi for suppression of essential genes. We set out to directly target cell proliferation by screening an RNAi library against DNA replication factors and identified multiple shRNAs against Replication Protein A, subunit 3 (RPA3). We generated transgenic mice with TRE-driven Rpa3 shRNAs whose expression enforced a reversible cell cycle arrest. In adult mice, the block in cell proliferation caused rapid atrophy of the intestinal epithelium which led to weight loss and lethality within 8-11 d of shRNA induction. Upon shRNA withdrawal, villus atrophy and weight loss were fully reversible. Thus, shRpa3 transgenic mice provide an interesting tool to study tissue maintenance and regeneration. Overall, we have established a robust system that serves the purpose of temperature-sensitive alleles in other model organisms, enabling inducible and reversible suppression of essential genes in a mammalian system.


Subject(s)
Alleles , Cell Cycle/physiology , DNA Replication/physiology , RNA Interference , Response Elements/physiology , Animals , Female , Male , Mice , Mice, Transgenic , Replication Protein A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL