Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167454, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39122224

ABSTRACT

Increasing evidence indicated that neuroinflammation was involved in progression of Parkinson's disease (PD). Long noncoding RNAs (lncRNAs) played important roles in regulating inflammatory processes in multiple kinds of human diseases such as cancer diabetes, cardiomyopathy, and neurodegenerative disorders. The mechanisms by which lncRNAs regulated PD related inflammation and dopaminergic neuronal loss have not yet been fully elucidated. In current study, we intended to explore the function and potential mechanism of lncRNA KCNQ1 opposite strand/antisense transcript 1 (KCNQ1OT1) in regulating inflammasome activation in PD. Functional assays confirmed that knockdown of KCNQ1OT1 suppress microglial NLR family pyrin domain containing 3 (NLRP3) inflammasome activation and attenuated dopaminergic neuronal loss in PD model mice. As KCNQ1OT1 located in both cytoplasm and nucleus of microglia, we demonstrated that KCNQ1OT1 promoted microglial NLRP3 inflammasome activation by competitive binding with miR-186 in cytoplasm and inhibited pri-miR-186 mediated NLRP3 silencing through recruitment of DiGeorge syndrome critical region gene 8 (DGCR8) in nucleus, respectively. Our study found a novel lncRNA-pri-miRNA/mature miRNA-mRNA regulatory network in microglia mediated NLRP3 inflammasome activation and dopaminergic neuronal loss, provided further insights for the treatment of Parkinson's disease.

2.
Mol Cell ; 84(14): 2665-2681.e13, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38955180

ABSTRACT

During implantation, embryos undergo an unpolarized-to-polarized transition to initiate postimplantation morphogenesis. However, the underlying molecular mechanism is unknown. Here, we identify a transient transcriptional activation governing embryonic morphogenesis and pluripotency transition during implantation. In naive pluripotent embryonic stem cells (ESCs), which represent preimplantation embryos, we find that the microprocessor component DGCR8 can recognize stem-loop structures within nascent mRNAs to sequester transcriptional coactivator FLII to suppress transcription directly. When mESCs exit from naive pluripotency, the ERK/RSK/P70S6K pathway rapidly activates, leading to FLII phosphorylation and disruption of DGCR8/FLII interaction. Phosphorylated FLII can bind to transcription factor JUN, activating cell migration-related genes to establish poised pluripotency akin to implanting embryos. Resequestration of FLII by DGCR8 drives poised ESCs into formative pluripotency. In summary, we identify a DGCR8/FLII/JUN-mediated transient transcriptional activation mechanism. Disruption of this mechanism inhibits naive-poised-formative pluripotency transition and the corresponding unpolarized-to-polarized transition during embryo implantation, which are conserved in mice and humans.


Subject(s)
Embryo Implantation , Gene Expression Regulation, Developmental , Morphogenesis , Transcriptional Activation , Animals , Embryo Implantation/genetics , Mice , Humans , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Phosphorylation , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/cytology , Female , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-jun/genetics , Signal Transduction
3.
Cells ; 13(7)2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38607000

ABSTRACT

MicroRNAs (miRNAs) act as negative regulators for protein-coding gene expression impacting cell proliferation, differentiation, and survival. These miRNAs are frequently dysregulated in cancer and constitute classes of blood-based biomarkers useful for cancer detection and prognosis definition. In thyroid cancer (TC), the miRNA biogenesis pathway plays a pivotal role in thyroid gland formation, ensuring proper follicle development and hormone production. Several alterations in the miRNA biogenesis genes are reported as a causality for miRNA dysregulation. Mutations in microprocessor component genes are linked to an increased risk of developing TC; in particular, a recurrent mutation affecting DGCR8, the E518K. In this review, we explore these novel findings and resume the current state-of-the-art in miRNAs in thyroid carcinomas.


Subject(s)
MicroRNAs , Thyroid Neoplasms , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , Thyroid Neoplasms/genetics , Cell Differentiation , Mutation/genetics
4.
J Clin Endocrinol Metab ; 109(7): 1733-1744, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38252873

ABSTRACT

BACKGROUND: Mutations in micro-RNA (miRNA) regulators DICER1 and DGCR8 have recently been uncovered, revealing a potential novel mechanism driving thyroid tumor development. However, the true frequency of these hotspot mutations in follicular-patterned thyroid tumors (FTs) and their relation to established driver gene events remain elusive. METHODS: A total of 440 FTs from 2 institutions were interrogated for DICER1, DGCR8, and RAS family hotspot mutations using Sanger sequencing. Whole-exome sequencing was also performed to identify additional driver gene aberrations in DICER1/DGCR8-mutant cases. Subsets of cases were further analyzed using miRNA expression profiling, and key dysregulated miRNAs were validated as markers of DICER1 mutations using quantitative RT-PCR analysis. The Cancer Genome Atlas (TCGA) database was also probed for DICER1/DGCR8 mutations and miRNA dysregulation. RESULTS: Fourteen (3.2%) and 4 (1%) FTs harbored DICER1 and DGCR8 hotspot mutations, respectively, in the combined cohort, and no cases with normal tissue available were found to exhibit a constitutional variant. Two DGCR8-mutant cases also harbored oncogenic RAS mutations. Whole-exome sequencing analysis did not identify additional driver gene events in DICER1/DGCR8-positive cases. Comprehensive miRNA expression profiling revealed a unique pattern of dysregulated miRNAs in DICER1/DGCR8-mutant cases compared with wild-type lesions. Moreover, DICER1-mutant cases showed a remarkable reduction of 5' arm miRNAs, findings corroborated in the TCGA cohort. CONCLUSION: DICER1 and DGCR8 hotspot mutations are rare in unselected cohorts of FTs, and mutated cases exhibit a specific miRNA profile. Although DGCR8 mutations may coexist with established RAS gene alterations, FTs with DICER1 variants were devoid of other driver gene events.


Subject(s)
DEAD-box RNA Helicases , MicroRNAs , Mutation , RNA-Binding Proteins , Ribonuclease III , Thyroid Nodule , Humans , Ribonuclease III/genetics , Female , DEAD-box RNA Helicases/genetics , Male , RNA-Binding Proteins/genetics , Middle Aged , Adult , MicroRNAs/genetics , Thyroid Nodule/genetics , Thyroid Nodule/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Aged , Adenocarcinoma, Follicular/genetics , Adenocarcinoma, Follicular/pathology , Prevalence , Exome Sequencing , Gene Expression Regulation, Neoplastic
5.
Dev Biol ; 506: 72-84, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38110169

ABSTRACT

The DGCR8 gene, encoding a critical miRNA processing protein, maps within the hemizygous region in patients with 22q11.2 deletion syndrome. Most patients have malformations of the cardiac outflow tract that is derived in part from the anterior second heart field (aSHF) mesoderm. To understand the function of Dgcr8 in the aSHF, we inactivated it in mice using Mef2c-AHF-Cre. Inactivation resulted in a fully penetrant persistent truncus arteriosus and a hypoplastic right ventricle leading to lethality by E14.5. To understand the molecular mechanism for this phenotype, we performed gene expression profiling of the aSHF and the cardiac outflow tract with right ventricle in conditional null versus normal mouse littermates at stage E9.5 prior to morphology changes. We identified dysregulation of mRNA gene expression, of which some are relevant to cardiogenesis. Many pri-miRNA genes were strongly increased in expression in mutant embryos along with reduced expression of mature miRNA genes. We further examined the individual, mature miRNAs that were decreased in expression along with pri-miRNAs that were accumulated that could be direct effects due to loss of Dgcr8. Among these genes, were miR-1a, miR-133a, miR-134, miR143 and miR145a, which have known functions in heart development. These early mRNA and miRNA changes may in part, explain the first steps that lead to the resulting phenotype in Dgcr8 aSHF conditional mutant embryos.


Subject(s)
Heart Ventricles , MicroRNAs , Humans , Mice , Animals , Heart Ventricles/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Mammals/metabolism , RNA, Messenger
6.
Methods Enzymol ; 692: 217-230, 2023.
Article in English | MEDLINE | ID: mdl-37925180

ABSTRACT

The Microprocessor complex (MP) is a vital component in the biogenesis of microRNAs (miRNAs) in animals. It plays a crucial role in the biogenesis of microRNAs (miRNAs) in mammals as it cleaves primary miRNAs (pri-miRNAs) to initiate their production. The accurate enzymatic activity of MP is critical to ensuring proper sequencing and expression of miRNAs and their correct cellular functions. RNA elements in pri-miRNAs, including secondary structures and sequencing motifs, RNA editing and modifications, and cofactors, can impact MP cleavage and affect miRNA expression and sequence. To evaluate MP cleavage activity with various RNA substrates under different conditions, we set up an in vitro pri-miRNA cleavage assay. This involves purifying human MP from HEK293E cells, synthesizing pri-miRNAs using in vitro transcription, and performing pri-miRNA cleavage assays using basic laboratory equipment and reagents. These procedures can be performed in various labs and improved for high-throughput analysis of enzymatic activities with thousands of RNA substrates.


Subject(s)
MicroRNAs , RNA Processing, Post-Transcriptional , Animals , Humans , Ribonuclease III/chemistry , Ribonuclease III/genetics , Ribonuclease III/metabolism , MicroRNAs/chemistry , RNA Editing , Microcomputers , Mammals/genetics
7.
Mol Cell ; 83(11): 1810-1826.e8, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37267903

ABSTRACT

Microprocessor (MP), DROSHA-DGCR8, processes primary miRNA transcripts (pri-miRNAs) to initiate miRNA biogenesis. The canonical cleavage mechanism of MP has been extensively investigated and comprehensively validated for two decades. However, this canonical mechanism cannot account for the processing of certain pri-miRNAs in animals. In this study, by conducting high-throughput pri-miRNA cleavage assays for approximately 260,000 pri-miRNA sequences, we discovered and comprehensively characterized a noncanonical cleavage mechanism of MP. This noncanonical mechanism does not need several RNA and protein elements essential for the canonical mechanism; instead, it utilizes previously unrecognized DROSHA dsRNA recognition sites (DRESs). Interestingly, the noncanonical mechanism is conserved across animals and plays a particularly significant role in C. elegans. Our established noncanonical mechanism elucidates MP cleavage in numerous RNA substrates unaccounted for by the canonical mechanism in animals. This study suggests a broader substrate repertoire of animal MPs and an expanded regulatory landscape for miRNA biogenesis.


Subject(s)
MicroRNAs , Animals , MicroRNAs/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , RNA-Binding Proteins/metabolism , Ribonuclease III/metabolism , RNA, Double-Stranded , RNA Processing, Post-Transcriptional
8.
Viruses ; 15(5)2023 05 16.
Article in English | MEDLINE | ID: mdl-37243263

ABSTRACT

miRNAs, small non-coding RNAs that regulate gene expression, are involved in various pathological processes, including viral infections. Virus infections may interfere with the miRNA pathway through the inhibition of genes involved in miRNA biogenesis. A reduction in the number and the levels of miRNAs expressed in nasopharyngeal swabs of patients with severe COVID-19 was lately observed by us, pointing towards the potential of miRNAs as possible diagnostic or prognostic biomarkers for predicting outcomes among patients with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. The objective of the present study was to investigate whether SARS-CoV-2 infection influences the expression levels of messenger RNAs (mRNAs) of key genes involved in miRNA biogenesis. mRNA levels of AGO2, DICER1, DGCR8, DROSHA, and Exportin-5 (XPO5) were measured by quantitative reverse-transcription polymerase chain reaction (RT-qPCR) in nasopharyngeal swab specimens from patients with COVID-19 and controls, as well as in cells infected with SARS-CoV-2 in vitro. Our data showed that the mRNA expression levels of AGO2, DICER1, DGCR8, DROSHA, and XPO5 were not significantly different in patients with severe COVID-19 when compared to patients with non-severe COVID-19 and controls. Similarly, the mRNA expression of these genes was not affected by SARS-CoV-2 infection in NHBE and Calu-3 cells. However, in Vero E6 cells, AGO2, DICER1, DGCR8, and XPO5 mRNA levels were slightly upregulated 24 h after infection with SARS-CoV-2. In conclusion, we did not find evidence for downregulation of mRNA levels of miRNA biogenesis genes during SARS-CoV-2 infection, neither ex vivo nor in vitro.


Subject(s)
COVID-19 , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , COVID-19/genetics , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , RNA-Binding Proteins/metabolism , RNA, Messenger/genetics , Ribonuclease III/genetics , Ribonuclease III/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Karyopherins/genetics
9.
Tohoku J Exp Med ; 260(3): 193-204, 2023 Jul 08.
Article in English | MEDLINE | ID: mdl-37045786

ABSTRACT

Oral squamous cell carcinoma (OSCC) remains the most prevalent malignance in the head and neck with highly aggressive attributes. This study investigates the functions of nuclear receptor interacting protein 1 (NRIP1) and its target transcripts in the progression of OSCC. By analyzing four OSCC-related Gene Expression Omnibus (GEO) datasets (GSE9844, GSE23558, GSE25104 and GSE74530) and querying bioinformatics systems, we obtained NRIP1 as an aberrantly highly expressed transcription factor in OSCC. Increased NRIP1 was detected in OSCC cell lines. Artificial downregulation of NRIP1 significantly suppressed proliferation, migration and invasion, resistance to apoptosis, tumorigenicity, and in vivo metastatic potential of OSCC cells. Moreover, the bioinformatics analyses suggested nuclear receptor binding SET domain protein 2 (NSD2) as a target of NRIP1 and DGCR8 microprocessor complex subunit (DGCR8) as a target of NSD2. Indeed, we validated by chromatin immunoprecipitation and luciferase assays that NRIP1 activated the transcription of NSD2, and NSD2 increased DGCR8 transcription by modulating histone methylation near the DGCR8 promoter. Either NSD2 or DGCR8 upregulation in OSCC cells rescued their malignant properties. Collectively, this study demonstrates that NRIP1 augments malignant properties of OSCC cells by activating NSD2-mediated histone methylation of DGCR8.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , MicroRNAs , Mouth Neoplasms , Humans , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Squamous Cell Carcinoma of Head and Neck/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , MicroRNAs/genetics , Histones/genetics , Histones/metabolism , Nuclear Receptor Interacting Protein 1/genetics , Nuclear Receptor Interacting Protein 1/metabolism , RNA-Binding Proteins/metabolism , DNA Methylation , Head and Neck Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Cell Movement/genetics
10.
Trends Genet ; 39(5): 401-414, 2023 05.
Article in English | MEDLINE | ID: mdl-36863945

ABSTRACT

MicroRNAs (miRNAs) play vital roles in the regulation of gene expression, a process known as miRNA-induced gene silencing. The human genome codes for many miRNAs, and their biogenesis relies on a handful of genes, including DROSHA, DGCR8, DICER1, and AGO1/2. Germline pathogenic variants (GPVs) in these genes cause at least three distinct genetic syndromes, with clinical manifestations that range from hyperplastic/neoplastic entities to neurodevelopmental disorders (NDDs). Over the past decade, DICER1 GPVs have been shown to lead to tumor predisposition. Moreover, recent findings have provided insight into the clinical consequences arising from GPVs in DGCR8, AGO1, and AGO2. Here we provide a timely update with respect to how GPVs in miRNA biogenesis genes alter miRNA biology and ultimately lead to their clinical manifestations.


Subject(s)
MicroRNAs , Humans , MicroRNAs/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Genotype , Genome, Human , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism
11.
Int J Mol Sci ; 23(23)2022 Nov 26.
Article in English | MEDLINE | ID: mdl-36499151

ABSTRACT

DGCR8 emerged recently as miRNAs biogenesis pathway protein with a highlighted role in thyroid disease. This study aimed to characterize this miRNA biogenesis component, in particular the p.(E518K) mutation and DGCR8 expression in a series of thyroid lesions. The series of thyroid lesions was genotyped for the c.1552G>A p.(E518K) mutation. When frozen tissue was available, DGCR8 mRNA expression was analysed by qPCR. Formalin-fixed paraffin-embedded tissues were studied for DGCR8 immunoexpression. We present for the first time the p.(E518K) mutation in a case of poorly differentiated thyroid carcinoma and present the deregulation of DGCR8 expression at mRNA level in follicular-patterned tumours. The obtained data solidify DGCR8 as another important player of miRNA-related gene mutations in thyroid tumorigenesis, particularly in follicular-patterned thyroid tumours.


Subject(s)
MicroRNAs , Neoplasms , Humans , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Thyroid Gland/metabolism , RNA, Messenger , Mutation
12.
Proc Natl Acad Sci U S A ; 119(45): e2212942119, 2022 Nov 08.
Article in English | MEDLINE | ID: mdl-36322738

ABSTRACT

MicroRNAs (miRNAs) are small, noncoding RNAs that regulate gene expression after transcription. miRNAs are present in transcriptionally quiescent full-grown oocytes and preimplantation embryos that display a low level of transcription prior to embryonic genome activation. The role of miRNAs, if any, in preimplantation development is not known. The temporal pattern of expression of miRNAs during bovine preimplantation development was determined by small RNA-sequencing using eggs and preimplantation embryos (1-cell, 2-cell, 4-cell, 8-cell, 16-cell, morula, and blastocyst). Embryos cultured in the presence of α-amanitin, which permitted the distinguishing of maternal miRNAs from embryonic miRNAs, indicated that embryonic miRNA expression was first detected at the two-cell stage but dramatically increased during the morula and blastocyst stages. Targeting DGCR8 by a small-interfering RNA/morpholino approach revealed a role for miRNAs in the morula-to-blastocyst transition. Knockdown of DGCR8 not only inhibited expression of embryonically expressed miRNAs but also inhibited the morula-to-blastocyst transition. In addition, RNA-sequencing identified an increased relative abundance of messenger RNAs potentially targeted by embryonic miRNAs in DGCR8-knockdown embryos when compared with controls. Results from these experiments implicate an essential role for miRNAs in bovine preimplantation embryo development.


Subject(s)
MicroRNAs , RNA, Small Untranslated , Pregnancy , Female , Cattle , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Gene Expression Regulation, Developmental , RNA-Binding Proteins/metabolism , Embryonic Development/genetics , Blastocyst/metabolism , RNA, Small Untranslated/metabolism
13.
Hum Mutat ; 43(10): 1368-1376, 2022 10.
Article in English | MEDLINE | ID: mdl-35723634

ABSTRACT

Schwannomatosis comprises a group of hereditary tumor predisposition syndromes characterized by, usually benign, multiple nerve sheath tumors, which frequently cause severe pain that does not typically respond to drug treatments. The most common schwannomatosis-associated gene is NF2, but SMARCB1 and LZTR1 are also associated. There are still many cases in which no pathogenic variants (PVs) have been identified, suggesting the existence of as yet unidentified genetic risk factors. In this study, we performed extended genetic screening of 75 unrelated schwannomatosis patients without identified germline PVs in NF2, LZTR1, or SMARCB1. Screening of the coding region of DGCR8, COQ6, CDKN2A, and CDKN2B was carried out, based on previous reports that point to these genes as potential candidate genes for schwannomatosis. Deletions or duplications in CDKN2A, CDKN2B, and adjacent chromosome 9 region were assessed by multiplex ligation-dependent probe amplification analysis. Sequencing analysis of a patient with multiple schwannomas and melanomas identified a novel duplication in the coding region of CDKN2A, disrupting both p14ARF and p16INK4a. Our results suggest that none of these genes are major contributors to schwannomatosis risk but the possibility remains that they may have a role in more complex mechanisms for tumor predisposition.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16 , Neurilemmoma , Neurofibromatoses , Skin Neoplasms , Cyclin-Dependent Kinase Inhibitor p16/genetics , Humans , Neurilemmoma/genetics , Neurilemmoma/pathology , Neurofibromatoses/genetics , RNA-Binding Proteins , SMARCB1 Protein/genetics , Skin Neoplasms/genetics , Transcription Factors/genetics
14.
Front Psychiatry ; 13: 873873, 2022.
Article in English | MEDLINE | ID: mdl-35492695

ABSTRACT

DiGeorge Syndrome Critical Region Gene 8 (DGCR8) is a key component of the microprocessor complex governing the maturation of most microRNAs, some of which participate in schizophrenia and neural development. Previous studies have found that the 22q11.2 locus, containing DGCR8, confers a risk of schizophrenia. However, the role of DGCR8 in schizophrenia and the early stage of neural development has remained unknown. In the present study, we try to identify the role of DGCR8 in schizophrenia from human samples and animal models. We found that the G allele and GG genotype of rs3757 in DGCR8 conferred a higher risk of schizophrenia, which likely resulted from higher expression of DGCR8 according to our test of dual-luciferase reporter system. Employed overexpression model in utero and adult mice, we also revealed that the aberrant increase of Dgcr8 delayed neuronal migration during embryological development and consequently triggered abnormal behaviors in adult mice. Together, these results demonstrate that DGCR8 may play a role in the etiology of schizophrenia through regulating neural development.

15.
Front Genet ; 13: 812183, 2022.
Article in English | MEDLINE | ID: mdl-35495153

ABSTRACT

The 22q11.2 deletion is associated with >20-fold increased risk for schizophrenia. The presence of gene DGCR8 in the 22q11.2 deletion region has suggested microRNA (miRNA) dysregulation as possibly contributing to this risk. We therefore investigated the role of miRNA target genes in the context of previously identified genome-wide risk for schizophrenia conveyed by additional copy number variation (CNV) in 22q11.2 deletion syndrome (22q11.2DS). Using a cohort of individuals with 22q11.2DS and documented additional rare CNVs overlapping protein coding genes, we compared those with schizophrenia (n = 100) to those with no psychotic illness (n = 118), assessing for rare CNVs that overlapped experimentally supported miRNA target genes. We further characterized the contributing miRNA target genes using gene set enrichment analyses and identified the miRNAs most implicated. Consistent with our hypothesis, we found a significantly higher proportion of individuals in the schizophrenia than in the non-psychotic group to have an additional rare CNV that overlapped one or more miRNA target genes (odds ratio = 2.12, p = 0.0138). Gene set analyses identified an enrichment of FMRP targets and genes involved in nervous system development and postsynaptic density amongst these miRNA target genes in the schizophrenia group. The miRNAs most implicated included miR-17-5p, miR-34a-5p and miR-124-3p. These results provide initial correlational evidence in support of a possible role for miRNA perturbation involving genes affected by rare genome-wide CNVs in the elevated risk for schizophrenia in 22q11.2DS, consistent with the multi-hit and multi-layered genetic mechanisms implicated in this and other forms of schizophrenia.

16.
Noncoding RNA ; 8(1)2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35202084

ABSTRACT

Micro-RNAs (miRNAs) are a class of non-coding RNAs (ncRNAs) that act as post-transcriptional regulators of gene expression. Since their discovery in 1993, they have been the subject of deep study due to their involvement in many important biological processes. Compared with other ncRNAs, miRNAs are generated from devoted transcriptional units which are processed by a specific set of endonucleases. The contribution of structural biology methods for understanding miRNA biogenesis and function has been essential for the dissection of their roles in cell biology and human disease. In this review, we summarize the application of structural biology for the characterization of the molecular players involved in miRNA biogenesis (processors and effectors), starting from the X-ray crystallography methods to the more recent cryo-electron microscopy protocols.

17.
Front Immunol ; 13: 991347, 2022.
Article in English | MEDLINE | ID: mdl-36591274

ABSTRACT

We have previously shown that the microRNA (miRNA) processor complex consisting of the RNAse Drosha and the DiGeorge Critical Region (DGCR) 8 protein is essential for B cell maturation. To determine whether miRNA processing is required to initiate T cell-mediated antibody responses, we deleted DGCR8 in maturing B2 cells by crossing a mouse with loxP-flanked DGCR8 alleles with a CD23-Cre mouse. As expected, non-immunized mice showed reduced numbers of mature B2 cells and IgG-secreting cells and diminished serum IgG titers. In accordance, germinal centers and antigen-specific IgG-secreting cells were absent in mice immunized with T-dependent antigens. Therefore, DGCR8 is required to mount an efficient T-dependent antibody response. However, DGCR8 deletion in B1 cells was incomplete, resulting in unaltered B1 cell numbers and normal IgM and IgA titers in DGCR8-knock-out mice. Therefore, this mouse model could be used to analyze B1 responses in the absence of functional B2 cells.


Subject(s)
MicroRNAs , Mice , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , T-Lymphocytes/metabolism , Germinal Center/metabolism , Immunoglobulin G/metabolism
18.
Curr Biol ; 32(1): 14-25.e4, 2022 01 10.
Article in English | MEDLINE | ID: mdl-34678162

ABSTRACT

Schizophrenia results from hundreds of known causes, including genetic, environmental, and developmental insults that cooperatively increase risk of developing the disease. In spite of the diversity of causal factors, schizophrenia presents with a core set of symptoms and brain abnormalities (both structural and functional) that particularly impact the prefrontal cortex. This suggests that many different causal factors leading to schizophrenia may cause prefrontal neurons and circuits to fail in fundamentally similar ways. The nature of convergent malfunctions in prefrontal circuits at the cell and synaptic levels leading to schizophrenia are not known. Here, we apply convergence-guided search to identify core pathological changes in the functional properties of prefrontal circuits that lie downstream of mechanistically distinct insults relevant to the disease. We compare the impacts of blocking NMDA receptors in monkeys and deleting a schizophrenia risk gene in mice on activity timing and effective communication in prefrontal local circuits. Although these manipulations operate through distinct molecular pathways and biological mechanisms, we found they produced convergent pathophysiological effects on prefrontal local circuits. Both manipulations reduced the frequency of synchronous (0-lag) spiking between prefrontal neurons and weakened functional interactions between prefrontal neurons at monosynaptic lags as measured by information transfer between the neurons. The two observations may be related, as reduction in synchronous spiking between prefrontal neurons would be expected to weaken synaptic connections between them via spike-timing-dependent synaptic plasticity. These data suggest that the link between spike timing and synaptic connectivity could comprise the functional vulnerability that multiple risk factors exploit to produce disease.


Subject(s)
Schizophrenia , Animals , Mice , Neurons/metabolism , Prefrontal Cortex/physiology , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia/genetics
19.
FEBS Lett ; 595(23): 2897-2908, 2021 12.
Article in English | MEDLINE | ID: mdl-34778960

ABSTRACT

Cellular double-stranded RNA-binding proteins (DRBPs) play important roles in the regulation of innate immune responses and microRNA (miRNA) biogenesis. The current study aimed to understand whether OV20.0, a DRBP of orf virus (ORFV), is involved in cellular RNA biogenesis via association with host DRBPs. We found that OV20.0 interacts with DiGeorge syndrome critical region 8 (DGCR8), a subunit of the miRNA processor complex, and binds to primary- and precursor-miRNA. Additionally, OV20.0 regulates DGCR8 expression in multiple ways, including through interaction with the DGCR8 protein and binding to DGCR8 mRNA. Lastly, our data show that DGCR8 plays an antiviral role against ORFV infection, whereas it is beneficial for influenza virus propagation, indicating that the underlying mechanisms could be diverse among different viruses.


Subject(s)
Ecthyma, Contagious/virology , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , Viral Proteins/metabolism , A549 Cells , Animals , Dogs , Ecthyma, Contagious/metabolism , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , MicroRNAs/genetics , Orf virus/pathogenicity , Protein Binding , RNA, Messenger/metabolism
20.
Eur J Neurosci ; 54(10): 7476-7492, 2021 11.
Article in English | MEDLINE | ID: mdl-34735028

ABSTRACT

The current study is focused on mechanisms by which the peripheral circadian oscillator in the prefrontal cortex (PFC) participates in food reward-induced activity. The experimental group of male Wistar rats was trained to receive a food reward with a low hedonic and caloric value. Afterwards, animals were exposed to a 5 h phase advance. Experimental animals could access a small food reward as they had been accustomed to, while control rats were exposed to the same phase shift without access to a food reward. When synchronisation to a new light:dark cycle was accompanied by intake of food reward, animals exerted more exact phase shift compared to the controls. In rats with access to a food reward, a rhythm in dopamine receptors types 1 and 2 in the PFC was detected. Rhythmic clock gene expression was induced in the PFC of rats when a food reward was provided together with a phase shift. The per2 and clock genes are predicted targets of miR-34a-5p. The precursor form of miR-34a-5p (pre-miR-34a-5p) showed a daily rhythm in expression in the PFC of the control and experimental groups. On the other hand, the mature form of miR-34a-5p exerted an inverted rhythm compared to pre-miR-34a-5p and negative correlation with per and clock genes expression only in the PFC of rewarded rats. A difference in the pattern of mature and pre-miR-34a-5p values was not related to expression of enzymes drosha, dicer and dgcr8. A role of the clock genes and miR-34a-5p in reward-facilitated synchronisation has been hypothesised.


Subject(s)
MicroRNAs , Animals , Gene Expression , Male , MicroRNAs/genetics , Prefrontal Cortex , RNA-Binding Proteins , Rats , Rats, Wistar , Reward
SELECTION OF CITATIONS
SEARCH DETAIL