Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(10): e2308255121, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38412125

RESUMO

MicroRNAs (miRNA) associate with Argonaute (AGO) proteins and repress gene expression by base pairing to sequences in the 3' untranslated regions of target genes. De novo coding variants in the human AGO genes AGO1 and AGO2 cause neurodevelopmental disorders (NDD) with intellectual disability, referred to as Argonaute syndromes. Most of the altered amino acids are conserved between the miRNA-associated AGO in Homo sapiens and Caenorhabditis elegans, suggesting that the human mutations could disrupt conserved functions in miRNA biogenesis or activity. We genetically modeled four human AGO1 mutations in C. elegans by introducing identical mutations into the C. elegans AGO1 homologous gene, alg-1. These alg-1 NDD mutations cause phenotypes in C. elegans indicative of disrupted miRNA processing, miRISC (miRNA silencing complex) formation, and/or target repression. We show that the alg-1 NDD mutations are antimorphic, causing developmental and molecular phenotypes stronger than those of alg-1 null mutants, likely by sequestrating functional miRISC components into non-functional complexes. The alg-1 NDD mutations cause allele-specific disruptions in mature miRNA profiles, accompanied by perturbation of downstream gene expression, including altered translational efficiency and/or messenger RNA abundance. The perturbed genes include those with human orthologs whose dysfunction is associated with NDD. These cross-clade genetic studies illuminate fundamental AGO functions and provide insights into the conservation of miRNA-mediated post-transcriptional regulatory mechanisms.


Assuntos
Proteínas de Caenorhabditis elegans , MicroRNAs , Transtornos do Neurodesenvolvimento , Animais , Humanos , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , MicroRNAs/metabolismo , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Mutação
2.
Genes Dev ; 31(4): 422-437, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28279983

RESUMO

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.


Assuntos
Caenorhabditis elegans/embriologia , Caenorhabditis elegans/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , MicroRNAs/metabolismo , Processos de Determinação Sexual/genética , Animais , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Feminino , Masculino , MicroRNAs/genética , Mutação , Proteínas de Ligação a RNA/metabolismo
3.
Cell ; 136(5): 926-38, 2009 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-19269369

RESUMO

TRIM-NHL proteins represent a large class of metazoan proteins implicated in development and disease. We demonstrate that a C. elegans TRIM-NHL protein, NHL-2, functions as a cofactor for the microRNA-induced silencing complex (miRISC) and thereby enhances the posttranscriptional repression of several genetically verified microRNA targets, including hbl-1 and let-60/Ras (by the let-7 family of microRNAs) and cog-1 (by the lsy-6 microRNA). NHL-2 is localized to cytoplasmic P-bodies and physically associates with the P-body protein CGH-1 and the core miRISC components ALG-1/2 and AIN-1. nhl-2 and cgh-1 mutations compromise the repression of microRNA targets in vivo but do not affect microRNA biogenesis, indicating a role for an NHL-2:CGH-1 complex in the effector phase of miRISC activity. We propose that the NHL-2:CGH-1 complex functions in association with mature miRISC to modulate the efficacy of microRNA:target interactions in response to physiological and developmental signals, thereby ensuring the robustness of genetic regulatory pathways regulated by microRNAs.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/metabolismo , Proteínas de Transporte/metabolismo , MicroRNAs/metabolismo , RNA de Helmintos/metabolismo , Animais , Proteínas de Caenorhabditis elegans/genética , Proteínas de Transporte/genética , Grânulos Citoplasmáticos/metabolismo , Proteínas de Homeodomínio/metabolismo , RNA Nucleotidiltransferases/genética , RNA Nucleotidiltransferases/metabolismo , Complexo de Inativação Induzido por RNA/metabolismo
4.
Postepy Biochem ; 70(1): 62-70, 2024 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-39016233

RESUMO

MicroRNAs are small RNAs that enable parts of the genome to regulate the other parts of the genome by RNA::RNA complementarity. Genes that encode microRNAs function as trans-acting regulators of hundreds of other genes, primarily by inhibiting the production of protein from mRNAs to which the microRNAs can bind by base pairing. MicroRNAs and their Argonaute partner proteins constitute a regulatory complex (the miRISC) that exhibits astonishing regulatory versatility. microRNAs have been shown to perform diverse roles in genetic regulatory networks (GRNs) - to control developmental switches, to dampen gene expression noise, to coordinate multigene functional modules, and more broadly, to confer robustness and resilience to developmental and homeostatic processes. Genetic analysis reveals that the function of particular microRNAs can be conditional, such that the microRNA is required under particular environmental or physiological conditions, but relatively dispensable under other conditions. The diversity and versatility of microRNA function in animal systems reflects the many ways that miRISC can be regulated by cellular signaling pathways, and the structure-function interplay among microRNA, target, and Argonaute.


Assuntos
Regulação da Expressão Gênica , MicroRNAs , MicroRNAs/metabolismo , MicroRNAs/genética , Animais , Redes Reguladoras de Genes , Proteínas Argonautas/metabolismo , Proteínas Argonautas/genética , Humanos
5.
PLoS Biol ; 18(12): e3000969, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33259473

RESUMO

Pathogens such as Pseudomonas aeruginosa advantageously modify animal host physiology, for example, by inhibiting host protein synthesis. Translational inhibition of insects and mammalian hosts by P. aeruginosa utilizes the well-known exotoxin A effector. However, for the infection of Caenorhabditis elegans by P. aeruginosa, the precise pathways and mechanism(s) of translational inhibition are not well understood. We found that upon exposure to P. aeruginosa PA14, C. elegans undergoes a rapid loss of intact ribosomes accompanied by the accumulation of ribosomes cleaved at helix 69 (H69) of the 26S ribosomal RNA (rRNA), a key part of ribosome decoding center. H69 cleavage is elicited by certain virulent P. aeruginosa isolates in a quorum sensing (QS)-dependent manner and independently of exotoxin A-mediated translational repression. H69 cleavage is antagonized by the 3 major host defense pathways defined by the pmk-1, fshr-1, and zip-2 genes. The level of H69 cleavage increases with the bacterial exposure time, and it is predominantly localized in the worm's intestinal tissue. Genetic and genomic analysis suggests that H69 cleavage leads to the activation of the worm's zip-2-mediated defense response pathway, consistent with translational inhibition. Taken together, our observations suggest that P. aeruginosa deploys a virulence mechanism to induce ribosome degradation and H69 cleavage of host ribosomes. In this manner, P. aeruginosa would impair host translation and block antibacterial responses.


Assuntos
Infecções por Pseudomonas/genética , Pseudomonas aeruginosa/metabolismo , RNA Ribossômico/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina Básica , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/microbiologia , Proteínas de Caenorhabditis elegans/metabolismo , Citocinese/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata/imunologia , Proteínas Quinases Ativadas por Mitógeno , Biossíntese de Proteínas/genética , Biossíntese de Proteínas/fisiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/patogenicidade , Percepção de Quorum/genética , RNA Ribossômico/genética , Ribossomos/genética , Ribossomos/metabolismo , Virulência , Fatores de Virulência/genética
6.
Proc Natl Acad Sci U S A ; 117(39): 24213-24223, 2020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-32929008

RESUMO

MicroRNAs (miRNAs) function cell-intrinsically to regulate gene expression by base-pairing to complementary mRNA targets while in association with Argonaute, the effector protein of the miRNA-mediated silencing complex (miRISC). A relatively dilute population of miRNAs can be found extracellularly in body fluids such as human blood plasma and cerebrospinal fluid (CSF). The remarkable stability of circulating miRNAs in such harsh extracellular environments can be attributed to their association with protective macromolecular complexes, including extracellular vesicles (EVs), proteins such as Argonaut 2 (AGO2), or high-density lipoproteins. The precise origins and the potential biological significance of various forms of miRNA-containing extracellular complexes are poorly understood. It is also not known whether extracellular miRNAs in their native state may retain the capacity for miRISC-mediated target RNA binding. To explore the potential functionality of circulating extracellular miRNAs, we comprehensively investigated the association between circulating miRNAs and the miRISC Argonaute AGO2. Using AGO2 immunoprecipitation (IP) followed by small-RNA sequencing, we find that miRNAs in circulation are primarily associated with antibody-accessible miRISC/AGO2 complexes. Moreover, we show that circulating miRNAs can base-pair with a target mimic in a seed-based manner, and that the target-bound AGO2 can be recovered from blood plasma in an ∼1:1 ratio with the respective miRNA. Our findings suggest that miRNAs in circulation are largely contained in functional miRISC/AGO2 complexes under normal physiological conditions. However, we find that, in human CSF, the assortment of certain extracellular miRNAs into free miRISC/AGO2 complexes can be affected by pathological conditions such as amyotrophic lateral sclerosis.


Assuntos
Proteínas Argonautas/sangue , MicroRNAs/sangue , Esclerose Lateral Amiotrófica/líquido cefalorraquidiano , Proteínas Argonautas/líquido cefalorraquidiano , Pareamento de Bases , Estudos de Casos e Controles , Humanos , Imunoprecipitação , MicroRNAs/líquido cefalorraquidiano
7.
Development ; 146(21)2019 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-31597658

RESUMO

MicroRNAs target complementary mRNAs for degradation or translational repression, reducing or preventing protein synthesis. In Caenorhabditis elegans, the transcription factor HBL-1 (Hunchback-like 1) promotes early larval (L2)-stage cell fates, and the let-7 family microRNAs temporally downregulate HBL-1 to enable the L2-to-L3 cell-fate progression. In parallel to let-7-family microRNAs, the conserved RNA-binding protein LIN-28 and its downstream gene lin-46 also act upstream of HBL-1 in regulating the L2-to-L3 cell-fate progression. The molecular function of LIN-46, and how the lin-28-lin-46 pathway regulates HBL-1, are not understood. Here, we report that the regulation of HBL-1 by the lin-28-lin-46 pathway is independent of the let-7/lin-4 microRNA complementary sites (LCSs) in the hbl-1 3'UTR, and involves stage-specific post-translational regulation of HBL-1 nuclear accumulation. We find that LIN-46 is necessary and sufficient to prevent nuclear accumulation of HBL-1. Our results illuminate that robust progression from L2 to L3 cell fates depends on the combination of two distinct modes of HBL-1 downregulation: decreased synthesis of HBL-1 via let-7-family microRNA activity, and decreased nuclear accumulation of HBL-1 via action of the lin-28-lin-46 pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Regiões 3' não Traduzidas , Alelos , Animais , Sistemas CRISPR-Cas , Caenorhabditis elegans , Diferenciação Celular , Linhagem da Célula , Progressão da Doença , Regulação para Baixo , Deleção de Genes , Fenótipo , Proteínas de Ligação a RNA/metabolismo
8.
Development ; 146(5)2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30745431

RESUMO

C. elegans heterochronic genes determine the timing of expression of specific cell fates in particular stages of developing larvae. However, their broader roles in coordinating developmental events across diverse tissues have been less well investigated. Here, we show that loss of lin-28, a central heterochronic regulator of hypodermal development, causes reduced fertility associated with abnormal somatic gonadal morphology. In particular, the abnormal spermatheca-uterine valve morphology of lin-28(lf) hermaphrodites traps embryos in the spermatheca, which disrupts ovulation and causes embryonic lethality. The same genes that act downstream of lin-28 in the regulation of hypodermal developmental timing also act downstream of lin-28 in somatic gonadal morphogenesis and fertility. Importantly, we find that hypodermal expression, but not somatic gonadal expression, of lin-28 is sufficient for restoring normal somatic gonadal morphology in lin-28(lf) mutants. We propose that the abnormal somatic gonadal morphogenesis of lin-28(lf) hermaphrodites results from temporal discoordination between the accelerated hypodermal development and normally timed somatic gonadal development. Thus, our findings exemplify how a cell-intrinsic developmental timing program can also control proper development of other interacting tissues, presumably by cell non-autonomous signal(s). This article has an associated 'The people behind the papers' interview.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/fisiologia , Gônadas/fisiologia , Morfogênese , Proteínas Repressoras/fisiologia , Animais , Animais Geneticamente Modificados , Diferenciação Celular , Movimento Celular , Feminino , Fertilização , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/fisiologia , Masculino , Mutação , Ovulação , Plasmídeos , Interferência de RNA , Proteínas de Ligação a RNA/fisiologia
9.
Development ; 146(5)2019 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-30770392

RESUMO

The sequence and roles in developmental progression of the microRNA let-7 are conserved. In general, transcription of the let-7 primary transcript (pri-let-7) occurs early in development, whereas processing of the mature let-7 microRNA arises during cellular differentiation. In Caenorhabditiselegans and other animals, the RNA-binding protein LIN-28 post-transcriptionally inhibits let-7 biogenesis at early developmental stages, but the mechanisms by which LIN-28 does this are not fully understood. Nor is it understood how the developmental regulation of let-7 might influence the expression or activities of other microRNAs of the same seed family. Here, we show that pri-let-7 is trans-spliced to the SL1 splice leader downstream of the let-7 precursor stem-loop, which produces a short polyadenylated downstream mRNA, and that this trans-splicing event negatively impacts the biogenesis of mature let-7 microRNA in cis Moreover, this trans-spliced mRNA contains sequences that are complementary to multiple members of the let-7 seed family (let-7fam) and negatively regulates let-7fam function in trans Thus, this study provides evidence for a mechanism by which splicing of a microRNA primary transcript can negatively regulate said microRNA in cis as well as other microRNAs in trans.


Assuntos
Caenorhabditis elegans/genética , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/genética , Trans-Splicing , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Microscopia de Fluorescência , Mutação , Fases de Leitura Aberta , RNA Mensageiro/metabolismo , Transcrição Gênica
10.
Mol Cell ; 56(3): 376-388, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25306920

RESUMO

Macroautophagy (autophagy) is a lysosome-dependent degradation process that has been implicated in age-associated diseases. Autophagy is involved in both cell survival and cell death, but little is known about the mechanisms that distinguish its use during these distinct cell fates. Here, we identify the microRNA miR-14 as being both necessary and sufficient for autophagy during developmentally regulated cell death in Drosophila. Loss of miR-14 prevented induction of autophagy during salivary gland cell death, but had no effect on starvation-induced autophagy in the fat body. Moreover, misexpression of miR-14 was sufficient to prematurely induce autophagy in salivary glands, but not in the fat body. Importantly, miR-14 regulates this context-specific autophagy through its target, inositol 1,4,5-trisphosphate kinase 2 (ip3k2), thereby affecting inositol 1,4,5-trisphosphate (IP3) signaling and calcium levels during salivary gland cell death. This study provides in vivo evidence of microRNA regulation of autophagy through modulation of IP3 signaling.


Assuntos
Autofagia , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , MicroRNAs/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Animais , Cálcio/metabolismo , Linhagem Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimologia , Drosophila melanogaster/crescimento & desenvolvimento , Inositol 1,4,5-Trifosfato/metabolismo , Larva/citologia , Larva/enzimologia , Larva/crescimento & desenvolvimento , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Interferência de RNA , Glândulas Salivares/citologia , Sistemas do Segundo Mensageiro
11.
Mol Cell ; 50(2): 281-7, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-23541767

RESUMO

In metazoans, microRNAs play a critical role in the posttranscriptional regulation of genes required for cell proliferation and differentiation. MicroRNAs themselves are regulated by a multitude of mechanisms influencing their transcription and posttranscriptional maturation. However, there is only sparse knowledge on pathways regulating the mature, functional form of microRNA. Here, we uncover the implication of the decapping scavenger protein DCS-1 in the control of microRNA turnover. In Caenorhabditis elegans, mutations in dcs-1 increase the levels of functional microRNAs. We demonstrate that DCS-1 interacts with the exonuclease XRN-1 to promote microRNA degradation in an independent manner from its known decapping scavenger activity, establishing two molecular functions for DCS-1. Our findings thus indicate that DCS-1 is part of a degradation complex that performs microRNA turnover in animals.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimologia , MicroRNAs/metabolismo , N-Glicosil Hidrolases/metabolismo , RNA de Helmintos/metabolismo , Animais , Proteínas de Caenorhabditis elegans/genética , Exorribonucleases/metabolismo , Expressão Gênica , Mutação , N-Glicosil Hidrolases/genética , Pirofosfatases , Interferência de RNA , Estabilidade de RNA , Complexo de Inativação Induzido por RNA/metabolismo
12.
Proc Natl Acad Sci U S A ; 112(18): E2366-75, 2015 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-25897023

RESUMO

Animals maintain their developmental robustness against natural stresses through numerous regulatory mechanisms, including the posttranscriptional regulation of gene expression by microRNAs (miRNAs). Caenorhabditis elegans miRNAs of the let-7 family (let-7-Fam) function semiredundantly to confer robust stage specificity of cell fates in the hypodermal seam cell lineages. Here, we show reciprocal regulatory interactions between let-7-Fam miRNAs and the innate immune response pathway in C. elegans. Upon infection of C. elegans larvae with the opportunistic human pathogen Pseudomonas aeruginosa, the developmental timing defects of certain let-7-Fam miRNA mutants are enhanced. This enhancement is mediated by the p38 MAPK innate immune pathway acting in opposition to let-7-Fam miRNA activity, possibly via the downstream Activating Transcription Factor-7 (ATF-7). Furthermore, let-7-Fam miRNAs appear to exert negative regulation on the worm's resistance to P. aeruginosa infection. Our results show that the inhibition of pathogen resistance by let-7 involves downstream heterochronic genes and the p38 MAPK pathway. These findings suggest that let-7-Fam miRNAs are integrated into innate immunity gene regulatory networks, such that this family of miRNAs modulates immune responses while also ensuring robust timing of developmental events under pathogen stress.


Assuntos
Caenorhabditis elegans/genética , Imunidade Inata , MicroRNAs/metabolismo , Alelos , Animais , Caenorhabditis elegans/imunologia , Linhagem da Célula , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Genes Reporter , Sistema Imunitário , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Mutação , Fenótipo , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa , Transdução de Sinais , Resultado do Tratamento , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Proc Natl Acad Sci U S A ; 112(3): E287-96, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25561544

RESUMO

Transitions between asymmetric (self-renewing) and symmetric (proliferative) cell divisions are robustly regulated in the context of normal development and tissue homeostasis. To genetically assess the regulation of these transitions, we used the postembryonic epithelial stem (seam) cell lineages of Caenorhabditis elegans. In these lineages, the timing of these transitions is regulated by the evolutionarily conserved heterochronic pathway, whereas cell division asymmetry is conferred by a pathway consisting of Wnt (Wingless) pathway components, including posterior pharynx defect (POP-1)/TCF, APC related/adenomatosis polyposis coli (APR-1)/APC, and LIT-1/NLK (loss of intestine/Nemo-like kinase). Here we explore the genetic regulatory mechanisms underlying stage-specific transitions between self-renewing and proliferative behavior in the seam cell lineages. We show that mutations of genes in the heterochronic developmental timing pathway, including lin-14 (lineage defect), lin-28, lin-46, and the lin-4 and let-7 (lethal defects)-family microRNAs, affect the activity of LIT-1/POP-1 cellular asymmetry machinery and APR-1 polarity during larval development. Surprisingly, heterochronic mutations that enhance LIT-1 activity in seam cells can simultaneously also enhance the opposing, POP-1 activity, suggesting a role in modulating the potency of the cellular polarizing activity of the LIT-1/POP-1 system as development proceeds. These findings illuminate how the evolutionarily conserved cellular asymmetry machinery can be coupled to microRNA-regulated developmental pathways for robust regulation of stem cell maintenance and proliferation during the course of development. Such genetic interactions between developmental timing regulators and cell polarity regulators could underlie transitions between asymmetric and symmetric stem cell fates in other systems and could be deregulated in the context of developmental disorders and cancer.


Assuntos
Caenorhabditis elegans/citologia , Diferenciação Celular , Células-Tronco/citologia , Animais , Caenorhabditis elegans/genética
14.
Proc Natl Acad Sci U S A ; 112(38): E5271-80, 2015 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-26351692

RESUMO

MicroRNAs are regulators of gene expression whose functions are critical for normal development and physiology. We have previously characterized mutations in a Caenorhabditis elegans microRNA-specific Argonaute ALG-1 (Argonaute-like gene) that are antimorphic [alg-1(anti)]. alg-1(anti) mutants have dramatically stronger microRNA-related phenotypes than animals with a complete loss of ALG-1. ALG-1(anti) miRISC (microRNA induced silencing complex) fails to undergo a functional transition from microRNA processing to target repression. To better understand this transition, we characterized the small RNA and protein populations associated with ALG-1(anti) complexes in vivo. We extensively characterized proteins associated with wild-type and mutant ALG-1 and found that the mutant ALG-1(anti) protein fails to interact with numerous miRISC cofactors, including proteins known to be necessary for target repression. In addition, alg-1(anti) mutants dramatically overaccumulated microRNA* (passenger) strands, and immunoprecipitated ALG-1(anti) complexes contained nonstoichiometric yields of mature microRNA and microRNA* strands, with some microRNA* strands present in the ALG-1(anti) Argonaute far in excess of the corresponding mature microRNAs. We show complex and microRNA-specific defects in microRNA strand selection and microRNA* strand disposal. For certain microRNAs (for example mir-58), microRNA guide strand selection by ALG-1(anti) appeared normal, but microRNA* strand release was inefficient. For other microRNAs (such as mir-2), both the microRNA and microRNA* strands were selected as guide by ALG-1(anti), indicating a defect in normal specificity of the strand choice. Our results suggest that wild-type ALG-1 complexes recognize structural features of particular microRNAs in the context of conducting the strand selection and microRNA* ejection steps of miRISC maturation.


Assuntos
Proteínas Argonautas/metabolismo , Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/genética , MicroRNAs/metabolismo , Proteínas de Ligação a RNA/fisiologia , Animais , Sequência de Bases , Northern Blotting , Proteínas de Caenorhabditis elegans/genética , DNA Complementar/metabolismo , Biblioteca Gênica , Espectrometria de Massas , Dados de Sequência Molecular , Mutação , Proteômica , Proteínas de Ligação a RNA/genética , Termodinâmica
16.
Nucleic Acids Res ; 43(1): e1, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25056322

RESUMO

The preparation and high-throughput sequencing of cDNA libraries from samples of small RNA is a powerful tool to quantify known small RNAs (such as microRNAs) and to discover novel RNA species. Interest in identifying the small RNA repertoire present in tissues and in biofluids has grown substantially with the findings that small RNAs can serve as indicators of biological conditions and disease states. Here we describe a novel and straightforward method to clone cDNA libraries from small quantities of input RNA. This method permits the generation of cDNA libraries from sub-picogram quantities of RNA robustly, efficiently and reproducibly. We demonstrate that the method provides a significant improvement in sensitivity compared to previous cloning methods while maintaining reproducible identification of diverse small RNA species. This method should have widespread applications in a variety of contexts, including biomarker discovery from scarce samples of human tissue or body fluids.


Assuntos
Clonagem Molecular/métodos , Biblioteca Gênica , MicroRNAs/sangue , Biotinilação , DNA Complementar/química , DNA Complementar/isolamento & purificação , Nucleotídeos de Desoxiuracil , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , MicroRNAs/química , MicroRNAs/isolamento & purificação , Isoformas de RNA/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
PLoS Genet ; 10(4): e1004286, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24763381

RESUMO

microRNAs function in diverse developmental and physiological processes by regulating target gene expression at the post-transcriptional level. ALG-1 is one of two Caenorhabditis elegans Argonautes (ALG-1 and ALG-2) that together are essential for microRNA biogenesis and function. Here, we report the identification of novel antimorphic (anti) alleles of ALG-1 as suppressors of lin-28(lf) precocious developmental phenotypes. The alg-1(anti) mutations broadly impair the function of many microRNAs and cause dosage-dependent phenotypes that are more severe than the complete loss of ALG-1. ALG-1(anti) mutant proteins are competent for promoting Dicer cleavage of microRNA precursors and for associating with and stabilizing microRNAs. However, our results suggest that ALG-1(anti) proteins may sequester microRNAs in immature and functionally deficient microRNA Induced Silencing Complexes (miRISCs), and hence compete with ALG-2 for access to functional microRNAs. Immunoprecipitation experiments show that ALG-1(anti) proteins display an increased association with Dicer and a decreased association with AIN-1/GW182. These findings suggest that alg-1(anti) mutations impair the ability of ALG-1 miRISC to execute a transition from Dicer-associated microRNA processing to AIN-1/GW182 associated effector function, and indicate an active role for ALG/Argonaute in mediating this transition.


Assuntos
Proteínas Argonautas/genética , Proteínas de Transporte/genética , MicroRNAs/genética , Mutação/genética , Alelos , Sequência de Aminoácidos , Animais , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Sequência Conservada , Regulação da Expressão Gênica no Desenvolvimento/genética , Dados de Sequência Molecular , Proteínas Repressoras/genética , Alinhamento de Sequência
18.
Proc Natl Acad Sci U S A ; 111(33): 12169-74, 2014 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-25092309

RESUMO

We have identified, by quantitative real-time PCR, hundreds of miRNAs that are dramatically elevated in the plasma or serum of acetaminophen (APAP) overdose patients. Most of these circulating microRNAs decrease toward normal levels during treatment with N-acetyl cysteine (NAC). We identified a set of 11 miRNAs whose profiles and dynamics in the circulation during NAC treatment can discriminate APAP hepatotoxicity from ischemic hepatitis. The elevation of certain miRNAs can precede the dramatic rise in the standard biomarker, alanine aminotransferase (ALT), and these miRNAs also respond more rapidly than ALT to successful treatment. Our results suggest that miRNAs can serve as sensitive diagnostic and prognostic clinical tools for severe liver injury and could be useful for monitoring drug-induced liver injury during drug discovery.


Assuntos
Acetaminofen/intoxicação , Acetilcisteína/uso terapêutico , Hepatite/sangue , Isquemia/sangue , MicroRNAs/sangue , Alanina Transaminase/sangue , Hepatite/complicações , Humanos , Intoxicação/sangue , Intoxicação/tratamento farmacológico , Reação em Cadeia da Polimerase em Tempo Real
19.
Anal Biochem ; 501: 66-74, 2016 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-26969789

RESUMO

MicroRNAs (miRNAs) are small noncoding RNA molecules that function in RNA silencing and posttranscriptional regulation of gene expression. miRNAs in biofluids are being used for clinical diagnosis as well as disease prediction. Efficient and reproducible isolation methods are crucial for extracellular RNA detection. To determine the best methodologies for miRNA detection from plasma, the performance of four RNA extraction kits, including an in-house kit, were determined with miScript miRNA assay technology; all were measured using a high-throughput quantitative polymerase chain reaction (qPCR) platform (BioMark System) with 90 human miRNA assays. In addition, the performances of complementary DNA (cDNA) and preamplification kits for TaqMan miRNA assays and miScript miRNA assays were compared using the same 90 miRNAs on the BioMark System. There were significant quantification cycle (Cq) value differences for the detection of miRNA targets between isolation kits. cDNA, preamplification, and qPCR performances were also varied. In summary, this study demonstrates differences among RNA isolation methods as measured by reverse transcription (RT)-qPCR. Importantly, differences were also noted in cDNA and preamplification performance using TaqMan and miScript. The in-house kit performed better than the other three kits. These findings demonstrate significant variability between isolation and detection methods for low-abundant miRNA detection from biofluids.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , MicroRNAs/sangue , MicroRNAs/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , DNA Complementar/genética , Feminino , Humanos , Masculino , MicroRNAs/genética
20.
Development ; 139(12): 2177-86, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22619389

RESUMO

In C. elegans larvae, the execution of stage-specific developmental events is controlled by heterochronic genes, which include those encoding a set of transcription factors and the microRNAs that regulate the timing of their expression. Under adverse environmental conditions, developing larvae enter a stress-resistant, quiescent stage called 'dauer'. Dauer larvae are characterized by the arrest of all progenitor cell lineages at a stage equivalent to the end of the second larval stage (L2). If dauer larvae encounter conditions favorable for resumption of reproductive growth, they recover and complete development normally, indicating that post-dauer larvae possess mechanisms to accommodate an indefinite period of interrupted development. For cells to progress to L3 cell fate, the transcription factor Hunchback-like-1 (HBL-1) must be downregulated. Here, we describe a quiescence-induced shift in the repertoire of microRNAs that regulate HBL-1. During continuous development, HBL-1 downregulation (and consequent cell fate progression) relies chiefly on three let-7 family microRNAs, whereas after quiescence, HBL-1 is downregulated primarily by the lin-4 microRNA in combination with an altered set of let-7 family microRNAs. We propose that this shift in microRNA regulation of HBL-1 expression involves an enhancement of the activity of lin-4 and let-7 microRNAs by miRISC modulatory proteins, including NHL-2 and LIN-46. These results illustrate how the employment of alternative genetic regulatory pathways can provide for the robust progression of progenitor cell fates in the face of temporary developmental quiescence.


Assuntos
Caenorhabditis elegans/citologia , Caenorhabditis elegans/genética , Linhagem da Célula/genética , Redes Reguladoras de Genes/genética , MicroRNAs/genética , Transdução de Sinais/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Transtornos do Desenvolvimento Sexual/genética , Regulação para Baixo/genética , Regulação da Expressão Gênica no Desenvolvimento , Larva/citologia , Larva/genética , MicroRNAs/metabolismo , Modelos Biológicos , Fenótipo , Complexo de Inativação Induzido por RNA/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA