Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
Cell ; 155(1): 121-34, 2013 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-24074865

RESUMO

The de novo DNA methyltransferase 3-like (Dnmt3L) is a catalytically inactive DNA methyltransferase that cooperates with Dnmt3a and Dnmt3b to methylate DNA. Dnmt3L is highly expressed in mouse embryonic stem cells (ESCs), but its function in these cells is unknown. Through genome-wide analysis of Dnmt3L knockdown in ESCs, we found that Dnmt3L is a positive regulator of methylation at the gene bodies of housekeeping genes and, more surprisingly, is also a negative regulator of methylation at promoters of bivalent genes. Dnmt3L is required for the differentiation of ESCs into primordial germ cells (PGCs) through the activation of the homeotic gene Rhox5. We demonstrate that Dnmt3L interacts with the Polycomb PRC2 complex in competition with the DNA methyltransferases Dnmt3a and Dnmt3b to maintain low methylation levels at the H3K27me3 regions. Thus, in ESCs, Dnmt3L counteracts the activity of de novo DNA methylases to maintain hypomethylation at promoters of bivalent developmental genes.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Células-Tronco Embrionárias/metabolismo , Regiões Promotoras Genéticas , Animais , Diferenciação Celular , DNA Metiltransferase 3A , Proteína Potenciadora do Homólogo 2 de Zeste , Células Germinativas/metabolismo , Histonas/metabolismo , Proteínas de Homeodomínio/genética , Camundongos , Complexo Repressor Polycomb 2/metabolismo , Proteínas do Grupo Polycomb/metabolismo , Fatores de Transcrição/genética , DNA Metiltransferase 3B
2.
EMBO J ; 39(13): e104163, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32484994

RESUMO

The relationships between impaired cortical development and consequent malformations in neurodevelopmental disorders, as well as the genes implicated in these processes, are not fully elucidated to date. In this study, we report six novel cases of patients affected by BBSOAS (Boonstra-Bosch-Schaff optic atrophy syndrome), a newly emerging rare neurodevelopmental disorder, caused by loss-of-function mutations of the transcriptional regulator NR2F1. Young patients with NR2F1 haploinsufficiency display mild to moderate intellectual disability and show reproducible polymicrogyria-like brain malformations in the parietal and occipital cortex. Using a recently established BBSOAS mouse model, we found that Nr2f1 regionally controls long-term self-renewal of neural progenitor cells via modulation of cell cycle genes and key cortical development master genes, such as Pax6. In the human fetal cortex, distinct NR2F1 expression levels encompass gyri and sulci and correlate with local degrees of neurogenic activity. In addition, reduced NR2F1 levels in cerebral organoids affect neurogenesis and PAX6 expression. We propose NR2F1 as an area-specific regulator of mouse and human brain morphology and a novel causative gene of abnormal gyrification.


Assuntos
Fator I de Transcrição COUP/metabolismo , Neocórtex/embriologia , Células-Tronco Neurais/metabolismo , Lobo Occipital/embriologia , Atrofias Ópticas Hereditárias/embriologia , Lobo Parietal/embriologia , Animais , Fator I de Transcrição COUP/genética , Modelos Animais de Doenças , Humanos , Camundongos , Neocórtex/patologia , Células-Tronco Neurais/patologia , Lobo Occipital/patologia , Atrofias Ópticas Hereditárias/genética , Atrofias Ópticas Hereditárias/patologia , Fator de Transcrição PAX6/genética , Fator de Transcrição PAX6/metabolismo , Lobo Parietal/patologia
3.
Nat Methods ; 18(3): 249-252, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33619392

RESUMO

RNA structure heterogeneity is a major challenge when querying RNA structures with chemical probing. We introduce DRACO, an algorithm for the deconvolution of coexisting RNA conformations from mutational profiling experiments. Analysis of the SARS-CoV-2 genome using dimethyl sulfate mutational profiling with sequencing (DMS-MaPseq) and DRACO, identifies multiple regions that fold into two mutually exclusive conformations, including a conserved structural switch in the 3' untranslated region. This work may open the way to dissecting the heterogeneity of the RNA structurome.


Assuntos
Algoritmos , Genoma Viral/genética , Conformação de Ácido Nucleico , RNA Viral/química , SARS-CoV-2/genética , Regiões 3' não Traduzidas/genética , COVID-19 , Humanos , Mutação/efeitos dos fármacos , Mutação/genética , RNA Viral/genética , Ésteres do Ácido Sulfúrico/farmacologia
5.
Phys Rev Lett ; 132(8): 080402, 2024 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-38457728

RESUMO

Quantum information scrambling is a unitary process that destroys local correlations and spreads information throughout the system, effectively hiding it in nonlocal degrees of freedom. In principle, unscrambling this information is possible with perfect knowledge of the unitary dynamics [B. Yoshida and A. Kitaev, arXiv:1710.03363.]. However, this Letter demonstrates that even without previous knowledge of the internal dynamics, information can be efficiently decoded from an unknown scrambler by monitoring the outgoing information of a local subsystem. We show that rapidly mixing but not fully chaotic scramblers can be decoded using Clifford decoders. The essential properties of a scrambling unitary can be efficiently recovered, even if the process is exponentially complex. Specifically, we establish that a unitary operator composed of t non-Clifford gates admits a Clifford decoder up to t≤n.

6.
Cell ; 138(6): 1122-36, 2009 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-19766566

RESUMO

The phosphorylation of the serine 10 at histone H3 has been shown to be important for transcriptional activation. Here, we report the molecular mechanism through which H3S10ph triggers transcript elongation of the FOSL1 gene. Serum stimulation induces the PIM1 kinase to phosphorylate the preacetylated histone H3 at the FOSL1 enhancer. The adaptor protein 14-3-3 binds the phosphorylated nucleosome and recruits the histone acetyltransferase MOF, which triggers the acetylation of histone H4 at lysine 16 (H4K16ac). This histone crosstalk generates the nucleosomal recognition code composed of H3K9acS10ph/H4K16ac determining a nucleosome platform for the bromodomain protein BRD4 binding. The recruitment of the positive transcription elongation factor b (P-TEFb) via BRD4 induces the release of the promoter-proximal paused RNA polymerase II and the increase of its processivity. Thus, the single phosphorylation H3S10ph at the FOSL1 enhancer triggers a cascade of events which activate transcriptional elongation.


Assuntos
Código das Histonas , Histonas/metabolismo , Transcrição Gênica , Animais , Linhagem Celular , Quinase 9 Dependente de Ciclina/metabolismo , Drosophila , Recuperação de Fluorescência Após Fotodegradação , Transferência Ressonante de Energia de Fluorescência , Humanos , Camundongos , Nucleossomos/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fatores de Transcrição/metabolismo , Leveduras
7.
Nucleic Acids Res ; 50(5): 2587-2602, 2022 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-35137201

RESUMO

The histone acetyltransferase p300 (also known as KAT3B) is a general transcriptional coactivator that introduces the H3K27ac mark on enhancers triggering their activation and gene transcription. Genome-wide screenings demonstrated that a large fraction of long non-coding RNAs (lncRNAs) plays a role in cellular processes and organ development although the underlying molecular mechanisms remain largely unclear (1,2). We found 122 lncRNAs that interacts directly with p300. In depth analysis of one of these, lncSmad7, is required to maintain ESC self-renewal and it interacts to the C-terminal domain of p300. lncSmad7 also contains predicted RNA-DNA Hoogsteen forming base pairing. Combined Chromatin Isolation by RNA precipitation followed by sequencing (ChIRP-seq) together with CRISPR/Cas9 mutagenesis of the target sites demonstrate that lncSmad7 binds and recruits p300 to enhancers in trans, to trigger enhancer acetylation and transcriptional activation of its target genes. Thus, these results unveil a new mechanism by which p300 is recruited to the genome.


Assuntos
Histonas , RNA Longo não Codificante , Acetilação , Acetiltransferases/metabolismo , Cromatina/genética , Elementos Facilitadores Genéticos , Histonas/genética , Histonas/metabolismo , RNA Longo não Codificante/metabolismo , Fatores de Transcrição de p300-CBP/genética , Fatores de Transcrição de p300-CBP/metabolismo
8.
EMBO J ; 38(3)2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30591554

RESUMO

Transcription factor TFEB is thought to control cellular functions-including in the vascular bed-primarily via regulation of lysosomal biogenesis and autophagic flux. Here, we report that TFEB also orchestrates a non-canonical program that controls the cell cycle/VEGFR2 pathway in the developing vasculature. In endothelial cells, TFEB depletion halts proliferation at the G1-S transition by inhibiting the CDK4/Rb pathway. TFEB-deficient cells attempt to compensate for this limitation by increasing VEGFR2 levels at the plasma membrane via microRNA-mediated mechanisms and controlled membrane trafficking. TFEB stimulates expression of the miR-15a/16-1 cluster, which limits VEGFR2 transcript stability and negatively modulates expression of MYO1C, a regulator of VEGFR2 trafficking to the cell surface. Altered levels of miR-15a/16-1 and MYO1C in TFEB-depleted cells cause increased expression of plasma membrane VEGFR2, but in a manner associated with low signaling strength. An endothelium-specific Tfeb-knockout mouse model displays defects in fetal and newborn mouse vasculature caused by reduced endothelial proliferation and by anomalous function of the VEGFR2 pathway. These previously unrecognized functions of TFEB expand its role beyond regulation of the autophagic pathway in the vascular system.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/fisiologia , Proliferação de Células , Embrião de Mamíferos/citologia , Endotélio Vascular/citologia , Regulação da Expressão Gênica no Desenvolvimento , Neovascularização Fisiológica , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Células Cultivadas , Embrião de Mamíferos/fisiologia , Endotélio Vascular/fisiologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
9.
FASEB J ; 36(7): e22401, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35726676

RESUMO

During skeletal myogenesis, the zinc-finger transcription factors SNAI1 and SNAI2, are expressed in proliferating myoblasts and regulate the transition to terminally differentiated myotubes while repressing pro-differentiation genes. Here, we demonstrate that SNAI1 is upregulated in vivo during the early phase of muscle regeneration induced by bupivacaine injury. Using shRNA-mediated gene silencing in C2C12 myoblasts and whole-transcriptome microarray analysis, we identified a collection of genes belonging to the endoplasmic reticulum (ER) stress pathway whose expression, induced by myogenic differentiation, was upregulated in absence of SNAI1. Among these, key ER stress genes, such as Atf3, Ddit3/Chop, Hspa5/Bip, and Fgf21, a myokine involved in muscle differentiation, were strongly upregulated. Furthermore, by promoter mutant analysis and Chromatin immune precipitation assay, we demonstrated that SNAI1 represses Fgf21 and Atf3 in proliferating myoblasts by directly binding to multiple E boxes in their respective promoter regions. Together, these data describe a new regulatory mechanism of myogenic differentiation involving the direct repressive action of SNAI1 on ER stress and Fgf21 expression, ultimately contributing to maintaining the proliferative and undifferentiated state of myoblasts.


Assuntos
Desenvolvimento Muscular , Fibras Musculares Esqueléticas , Fatores de Transcrição da Família Snail/metabolismo , Fator 3 Ativador da Transcrição/metabolismo , Diferenciação Celular , Linhagem Celular , Fatores de Crescimento de Fibroblastos , Desenvolvimento Muscular/genética , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/fisiologia , Regiões Promotoras Genéticas/genética , Regulação para Cima
10.
EMBO Rep ; 22(7): e50882, 2021 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-34085753

RESUMO

Injury in adult tissue generally reactivates developmental programs to foster regeneration, but it is not known whether this paradigm applies to growing tissue. Here, by employing blisters, we show that epidermal wounds heal at the expense of skin development. The regenerated epidermis suppresses the expression of tissue morphogenesis genes accompanied by delayed hair follicle (HF) growth. Lineage tracing experiments, cell proliferation dynamics, and mathematical modeling reveal that the progeny of HF junctional zone stem cells, which undergo a morphological transformation, repair the blisters while not promoting HF development. In contrast, the contribution of interfollicular stem cell progeny to blister healing is small. These findings demonstrate that HF development can be sacrificed for the sake of epidermal wound regeneration. Our study elucidates the key cellular mechanism of wound healing in skin blistering diseases.


Assuntos
Vesícula , Folículo Piloso , Adulto , Vesícula/genética , Células Epidérmicas , Epiderme , Humanos , Pele , Células-Tronco
11.
Nature ; 543(7643): 72-77, 2017 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-28225755

RESUMO

In mammals, DNA methylation occurs mainly at CpG dinucleotides. Methylation of the promoter suppresses gene expression, but the functional role of gene-body DNA methylation in highly expressed genes has yet to be clarified. Here we show that, in mouse embryonic stem cells, Dnmt3b-dependent intragenic DNA methylation protects the gene body from spurious RNA polymerase II entry and cryptic transcription initiation. Using different genome-wide approaches, we demonstrate that this Dnmt3b function is dependent on its enzymatic activity and recruitment to the gene body by H3K36me3. Furthermore, the spurious transcripts can either be degraded by the RNA exosome complex or capped, polyadenylated, and delivered to the ribosome to produce aberrant proteins. Elongating RNA polymerase II therefore triggers an epigenetic crosstalk mechanism that involves SetD2, H3K36me3, Dnmt3b and DNA methylation to ensure the fidelity of gene transcription initiation, with implications for intragenic hypomethylation in cancer.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA/genética , DNA/metabolismo , Genes/genética , RNA Mensageiro/biossíntese , Iniciação da Transcrição Genética , Animais , Linhagem Celular , DNA/química , DNA (Citosina-5-)-Metiltransferases/deficiência , DNA (Citosina-5-)-Metiltransferases/genética , Epigênese Genética , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/química , Histonas/metabolismo , Lisina/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Poliadenilação , Capuzes de RNA/metabolismo , RNA Polimerase II/metabolismo , Estabilidade de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ribossomos/metabolismo , Sítio de Iniciação de Transcrição , DNA Metiltransferase 3B
12.
BMC Biol ; 20(1): 171, 2022 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-35918713

RESUMO

BACKGROUND: The high-mobility group Hmga family of proteins are non-histone chromatin-interacting proteins which have been associated with a number of nuclear functions, including heterochromatin formation, replication, recombination, DNA repair, transcription, and formation of enhanceosomes. Due to its role based on dynamic interaction with chromatin, Hmga2 has a pathogenic role in diverse tumors and has been mainly studied in a cancer context; however, whether Hmga2 has similar physiological functions in normal cells remains less explored. Hmga2 was additionally shown to be required during the exit of embryonic stem cells (ESCs) from the ground state of pluripotency, to allow their transition into epiblast-like cells (EpiLCs), and here, we use that system to gain further understanding of normal Hmga2 function. RESULTS: We demonstrated that Hmga2 KO pluripotent stem cells fail to develop into EpiLCs. By using this experimental system, we studied the chromatin changes that take place upon the induction of EpiLCs and we observed that the loss of Hmga2 affects the histone mark H3K27me3, whose levels are higher in Hmga2 KO cells. Accordingly, a sustained expression of polycomb repressive complex 2 (PRC2), responsible for H3K27me3 deposition, was observed in KO cells. However, gene expression differences between differentiating wt vs Hmga2 KO cells did not show any significant enrichments of PRC2 targets. Similarly, endogenous Hmga2 association to chromatin in epiblast stem cells did not show any clear relationships with gene expression modification observed in Hmga2 KO. Hmga2 ChIP-seq confirmed that this protein preferentially binds to the chromatin regions associated with nuclear lamina. Starting from this observation, we demonstrated that nuclear lamina underwent severe alterations when Hmga2 KO or KD cells were induced to exit from the naïve state and this phenomenon is accompanied by a mislocalization of the heterochromatin mark H3K9me3 within the nucleus. As nuclear lamina (NL) is involved in the organization of 3D chromatin structure, we explored the possible effects of Hmga2 loss on this phenomenon. The analysis of Hi-C data in wt and Hmga2 KO cells allowed us to observe that inter-TAD (topologically associated domains) interactions in Hmga2 KO cells are different from those observed in wt cells. These differences clearly show a peculiar compartmentalization of inter-TAD interactions in chromatin regions associated or not to nuclear lamina. CONCLUSIONS: Overall, our results indicate that Hmga2 interacts with heterochromatic lamin-associated domains, and highlight a role for Hmga2 in the crosstalk between chromatin and nuclear lamina, affecting the establishment of inter-TAD interactions.


Assuntos
Membrana Nuclear , Células-Tronco Pluripotentes , Cromatina/genética , Cromatina/metabolismo , Proteína HMGA2/genética , Proteína HMGA2/metabolismo , Heterocromatina/metabolismo , Histonas/genética , Membrana Nuclear/metabolismo , Células-Tronco Pluripotentes/metabolismo , Complexo Repressor Polycomb 2/genética
13.
Phys Rev Lett ; 128(5): 050402, 2022 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-35179939

RESUMO

We introduce a novel measure for the quantum property of "nonstabilizerness"-commonly known as "magic"-by considering the Rényi entropy of the probability distribution associated to a pure quantum state given by the square of the expectation value of Pauli strings in that state. We show that this is a good measure of nonstabilizerness from the point of view of resource theory and show bounds with other known measures. The stabilizer Rényi entropy has the advantage of being easily computable because it does not need a minimization procedure. We present a protocol for an experimental measurement by randomized measurements. We show that the nonstabilizerness is intimately connected to out-of-time-order correlation functions and that maximal levels of nonstabilizerness are necessary for quantum chaos.

14.
Nucleic Acids Res ; 48(4): 1730-1747, 2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-31889184

RESUMO

Heterogeneity is a fundamental feature of complex phenotypes. So far, genomic screenings have profiled thousands of samples providing insights into the transcriptome of the cell. However, disentangling the heterogeneity of these transcriptomic Big Data to identify defective biological processes remains challenging. Here we present GSECA, a method exploiting the bimodal behavior of RNA-sequencing gene expression profiles to identify altered gene sets in heterogeneous patient cohorts. Using simulated and experimental RNA-sequencing data sets, we show that GSECA provides higher performances than other available algorithms in detecting truly altered biological processes in large cohorts. Applied to 5941 samples from 14 different cancer types, GSECA correctly identified the alteration of the PI3K/AKT signaling pathway driven by the somatic loss of PTEN and verified the emerging role of PTEN in modulating immune-related processes. In particular, we showed that, in prostate cancer, PTEN loss appears to establish an immunosuppressive tumor microenvironment through the activation of STAT3, and low PTEN expression levels have a detrimental impact on patient disease-free survival. GSECA is available at https://github.com/matteocereda/GSECA.


Assuntos
Big Data , Sequenciamento do Exoma/estatística & dados numéricos , RNA/genética , Transcriptoma/genética , Linhagem Celular Tumoral , Intervalo Livre de Doença , Regulação da Expressão Gênica/genética , Humanos , Internet , PTEN Fosfo-Hidrolase/genética , Fator de Transcrição STAT3/genética , Análise de Sequência de RNA , Transdução de Sinais/genética , Software , Microambiente Tumoral/genética
15.
Int J Mol Sci ; 23(11)2022 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-35682796

RESUMO

In the human brain, long non-coding RNAs (lncRNAs) are widely expressed in an exquisitely temporally and spatially regulated manner, thus suggesting their contribution to normal brain development and their probable involvement in the molecular pathology of neurodevelopmental disorders (NDD). Bypassing the classic protein-centric conception of disease mechanisms, some studies have been conducted to identify and characterize the putative roles of non-coding sequences in the genetic pathogenesis and diagnosis of complex diseases. However, their involvement in NDD, and more specifically in intellectual disability (ID), is still poorly documented and only a few genomic alterations affecting the lncRNAs function and/or expression have been causally linked to the disease endophenotype. Considering that a significant fraction of patients still lacks a genetic or molecular explanation, we expect that a deeper investigation of the non-coding genome will unravel novel pathogenic mechanisms, opening new translational opportunities. Here, we present evidence of the possible involvement of many lncRNAs in the etiology of different forms of ID and NDD, grouping the candidate disease-genes in the most frequently affected cellular processes in which ID-risk genes were previously collected. We also illustrate new approaches for the identification and prioritization of NDD-risk lncRNAs, together with the current strategies to exploit them in diagnosis.


Assuntos
Deficiência Intelectual , Transtornos do Neurodesenvolvimento , RNA Longo não Codificante , Genômica , Humanos , Deficiência Intelectual/genética , Transtornos do Neurodesenvolvimento/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
16.
Br J Haematol ; 193(3): 581-591, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33570193

RESUMO

Multiple functions of CD38 need exploring to expand clinical application of anti-CD38 antibodies in multiple myeloma (MM). We investigated membrane dynamics of MM cells and subsequent events when CD38 is targeted by therapeutic antibodies. Human MM cells (BF01) were co-cultured in vitro with therapeutic antibody (or control immunoglobulin G) and analysed using gene expression profiling. Microvesicles from antibody-exposed cells were analysed for differential gene and microRNA (miRNA) expression, and for phenotypic characterisation. Exposure of BF01 cells to anti-CD38 antibody resulted in CD38 membrane redistribution, upregulation of metabolism-related genes and downregulation of genes involved in cell cycle processes. Microvesicles derived from antibody-exposed cells showed increased CD73 and CD39 expression, presence of programmed death-ligand 1 and significant up-/down-modulation of miRNAs. Microvesicles accumulated around immunoglobulin Fc receptor-positive (FcR+ ) cells. Upon internalisation, natural killer cells displayed significantly increased expression of genes related to activation and immune response, and downregulation of genes involved in the cell cycle. Cells may use microvesicles to transmit signals distally as part of a survival strategy. Microvesicles are equipped on their surface with enzymatic machinery leading to production of tolerogenic adenosine. Further, they are internalised in FcR+ cells with significant functional modifications. These observations have relevance for improving anti-CD38 therapeutic antibodies through targeting this mechanism and its sequelae.


Assuntos
ADP-Ribosil Ciclase 1/biossíntese , Anticorpos Antineoplásicos/farmacologia , Membrana Celular/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicoproteínas de Membrana/biossíntese , Mieloma Múltiplo/metabolismo , Proteínas de Neoplasias/biossíntese , Linhagem Celular Tumoral , Humanos , MicroRNAs/biossíntese , Mieloma Múltiplo/tratamento farmacológico , RNA Neoplásico/biossíntese
17.
Nucleic Acids Res ; 47(13): 7003-7017, 2019 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-31053845

RESUMO

The influenza A virus (IAV) is a continuous health threat to humans as well as animals due to its recurring epidemics and pandemics. The IAV genome is segmented and the eight negative-sense viral RNAs (vRNAs) are transcribed into positive sense complementary RNAs (cRNAs) and viral messenger RNAs (mRNAs) inside infected host cells. A role for the secondary structure of IAV mRNAs has been hypothesized and debated for many years, but knowledge on the structure mRNAs adopt in vivo is currently missing. Here we solve, for the first time, the in vivo secondary structure of IAV mRNAs in living infected cells. We demonstrate that, compared to the in vitro refolded structure, in vivo IAV mRNAs are less structured but exhibit specific locally stable elements. Moreover, we show that the targeted disruption of these high-confidence structured domains results in an extraordinary attenuation of IAV replicative capacity. Collectively, our data provide the first comprehensive map of the in vivo structural landscape of IAV mRNAs, hence providing the means for the development of new RNA-targeted antivirals.


Assuntos
Vírus da Influenza A Subtipo H1N1/genética , RNA Mensageiro/química , Sequências Reguladoras de Ácido Nucleico , Algoritmos , Animais , Conjuntos de Dados como Assunto , Cães , Escherichia coli , Biblioteca Gênica , Modelos Moleculares , Conformação de Ácido Nucleico , RNA/química , Dobramento de RNA , RNA Antissenso , RNA Mensageiro/genética , Seleção Genética , Relação Estrutura-Atividade , Termodinâmica
18.
Int J Mol Sci ; 22(24)2021 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-34948395

RESUMO

Smad7 has been identified as a negative regulator of the transforming growth factor TGF-ß pathway by direct interaction with the TGF-ß type I receptor (TßR-I). Although Smad7 has also been shown to play TGF-ß unrelated functions in the cytoplasm and in the nucleus, a comprehensive analysis of its nuclear function has not yet been performed. Here, we show that in ESCs Smad7 is mainly nuclear and acts as a general transcription factor regulating several genes unrelated to the TGF-ß pathway. Loss of Smad7 results in the downregulation of several key stemness master regulators, including Pou5f1 and Zfp42, and in the upregulation of developmental genes, with consequent loss of the stem phenotype. Integrative analysis of genome-wide mapping data for Smad7 and ESC self-renewal and pluripotency transcriptional regulators revealed that Smad7 co-occupies promoters of highly expressed key stemness regulators genes, by binding to a specific consensus response element NCGGAAMM. Altogether, our data establishes Smad7 as a new, integral component of the regulatory circuitry that controls ESC identity.


Assuntos
Células-Tronco Embrionárias Murinas/metabolismo , Proteína Smad7/genética , Ativação Transcricional , Animais , Linhagem Celular , Proteínas de Ligação a DNA/genética , Regulação para Baixo , Deleção de Genes , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Proteínas Nucleares/genética , Fator 3 de Transcrição de Octâmero/genética , Receptor do Fator de Crescimento Transformador beta Tipo I/genética , Fator de Crescimento Transformador beta/genética
19.
Int J Mol Sci ; 22(13)2021 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-34201896

RESUMO

miR-29b2 and miR-29c play a suppressive role in breast cancer progression. C1orf132 (also named MIR29B2CHG) is the host gene for generating both microRNAs. However, the region also expresses longer transcripts with unknown functions. We employed bioinformatics and experimental approaches to decipher C1orf132 expression and function in breast cancer tissues. We also used the CRISPR/Cas9 technique to excise a predicted C1orf132 distal promoter and followed the behavior of the edited cells by real-time PCR, flow cytometry, migration assay, and RNA-seq techniques. We observed that C1orf132 long transcript is significantly downregulated in triple-negative breast cancer. We also identified a promoter for the longer transcripts of C1orf132 whose functionality was demonstrated by transfecting MCF7 cells with a C1orf132 promoter-GFP construct. Knocking-out the promoter by means of CRISPR/Cas9 revealed no alterations in the expression of the neighboring genes CD46 and CD34, while the expression of miR-29c was reduced by half. Furthermore, the promoter knockout elevated the migration ability of the edited cells. RNA sequencing revealed many up- and downregulated genes involved in various cellular pathways, including epithelial to mesenchymal transition and mammary gland development pathways. Altogether, we are reporting here the existence of an additional/distal promoter with an enhancer effect on miR-29 generation and an inhibitory effect on cell migration.


Assuntos
RNA Longo não Codificante/genética , Neoplasias de Mama Triplo Negativas/genética , Animais , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Regulação para Baixo , Transição Epitelial-Mesenquimal/genética , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Células MCF-7 , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Regiões Promotoras Genéticas , RNA Longo não Codificante/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo
20.
Entropy (Basel) ; 23(8)2021 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-34441214

RESUMO

We show that the most important measures of quantum chaos, such as frame potentials, scrambling, Loschmidt echo and out-of-time-order correlators (OTOCs), can be described by the unified framework of the isospectral twirling, namely the Haar average of a k-fold unitary channel. We show that such measures can then always be cast in the form of an expectation value of the isospectral twirling. In literature, quantum chaos is investigated sometimes through the spectrum and some other times through the eigenvectors of the Hamiltonian generating the dynamics. We show that thanks to this technique, we can interpolate smoothly between integrable Hamiltonians and quantum chaotic Hamiltonians. The isospectral twirling of Hamiltonians with eigenvector stabilizer states does not possess chaotic features, unlike those Hamiltonians whose eigenvectors are taken from the Haar measure. As an example, OTOCs obtained with Clifford resources decay to higher values compared with universal resources. By doping Hamiltonians with non-Clifford resources, we show a crossover in the OTOC behavior between a class of integrable models and quantum chaos. Moreover, exploiting random matrix theory, we show that these measures of quantum chaos clearly distinguish the finite time behavior of probes to quantum chaos corresponding to chaotic spectra given by the Gaussian Unitary Ensemble (GUE) from the integrable spectra given by Poisson distribution and the Gaussian Diagonal Ensemble (GDE).

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa