Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(36): e2120680119, 2022 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-35998224

RESUMO

The systemic immune response to viral infection is shaped by master transcription factors, such as NF-κB, STAT1, or PU.1. Although long noncoding RNAs (lncRNAs) have been suggested as important regulators of transcription factor activity, their contributions to the systemic immunopathologies observed during SARS-CoV-2 infection have remained unknown. Here, we employed a targeted single-cell RNA sequencing approach to reveal lncRNAs differentially expressed in blood leukocytes during severe COVID-19. Our results uncover the lncRNA PIRAT (PU.1-induced regulator of alarmin transcription) as a major PU.1 feedback-regulator in monocytes, governing the production of the alarmins S100A8/A9, key drivers of COVID-19 pathogenesis. Knockout and transgene expression, combined with chromatin-occupancy profiling, characterized PIRAT as a nuclear decoy RNA, keeping PU.1 from binding to alarmin promoters and promoting its binding to pseudogenes in naïve monocytes. NF-κB-dependent PIRAT down-regulation during COVID-19 consequently releases a transcriptional brake, fueling alarmin production. Alarmin expression is additionally enhanced by the up-regulation of the lncRNA LUCAT1, which promotes NF-κB-dependent gene expression at the expense of targets of the JAK-STAT pathway. Our results suggest a major role of nuclear noncoding RNA networks in systemic antiviral responses to SARS-CoV-2 in humans.


Assuntos
COVID-19 , Regulação da Expressão Gênica , Monócitos , RNA Longo não Codificante , SARS-CoV-2 , Alarminas/genética , COVID-19/genética , COVID-19/imunologia , Humanos , Janus Quinases/genética , Monócitos/imunologia , NF-kappa B/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA-Seq , SARS-CoV-2/imunologia , Fatores de Transcrição STAT/genética , Transdução de Sinais/genética , Análise de Célula Única
2.
Proc Natl Acad Sci U S A ; 117(16): 9042-9053, 2020 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-32241891

RESUMO

RNA has been proposed as an important scaffolding factor in the nucleus, aiding protein complex assembly in the dense intracellular milieu. Architectural contributions of RNA to cytosolic signaling pathways, however, remain largely unknown. Here, we devised a multidimensional gradient approach, which systematically locates RNA components within cellular protein networks. Among a subset of noncoding RNAs (ncRNAs) cosedimenting with the ubiquitin-proteasome system, our approach unveiled ncRNA MaIL1 as a critical structural component of the Toll-like receptor 4 (TLR4) immune signal transduction pathway. RNA affinity antisense purification-mass spectrometry (RAP-MS) revealed MaIL1 binding to optineurin (OPTN), a ubiquitin-adapter platforming TBK1 kinase. MaIL1 binding stabilized OPTN, and consequently, loss of MaIL1 blunted OPTN aggregation, TBK1-dependent IRF3 phosphorylation, and type I interferon (IFN) gene transcription downstream of TLR4. MaIL1 expression was elevated in patients with active pulmonary infection and was highly correlated with IFN levels in bronchoalveolar lavage fluid. Our study uncovers MaIL1 as an integral RNA component of the TLR4-TRIF pathway and predicts further RNAs to be required for assembly and progression of cytosolic signaling networks in mammalian cells.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Interferon Tipo I/genética , Proteínas de Membrana Transportadoras/metabolismo , RNA não Traduzido/metabolismo , Infecções Respiratórias/imunologia , Receptor 4 Toll-Like/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Adulto , Idoso , Buffy Coat/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Feminino , Regulação da Expressão Gênica/imunologia , Técnicas de Silenciamento de Genes , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/sangue , Interferon Tipo I/imunologia , Macrófagos , Masculino , Pessoa de Meia-Idade , Fosforilação/genética , Cultura Primária de Células , Proteínas Serina-Treonina Quinases/metabolismo , Estabilidade Proteica , RNA não Traduzido/sangue , RNA não Traduzido/genética , RNA-Seq , Infecções Respiratórias/sangue , Infecções Respiratórias/microbiologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Adulto Jovem
3.
Nucleic Acids Res ; 48(18): 10397-10412, 2020 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-32946572

RESUMO

The RNA helicase RIG-I plays a key role in sensing pathogen-derived RNA. Double-stranded RNA structures bearing 5'-tri- or diphosphates are commonly referred to as activating RIG-I ligands. However, endogenous RNA fragments generated during viral infection via RNase L also activate RIG-I. Of note, RNase-digested RNA fragments bear a 5'-hydroxyl group and a 2',3'-cyclic phosphate. How endogenous RNA fragments activate RIG-I despite the lack of 5'-phosphorylation has not been elucidated. Here we describe an endogenous RIG-I ligand (eRL) that is derived from the internal transcribed spacer 2 region (ITS2) of the 45S ribosomal RNA after partial RNase A digestion in vitro, RNase A protein transfection or RNase L activation. The immunostimulatory property of the eRL is dependent on 2',3'-cyclic phosphate and its sequence is characterized by a G-quadruplex containing sequence motif mediating guanosine-5'-triphosphate (GTP) binding. In summary, RNase generated self-RNA fragments with 2',3'-cyclic phosphate function as nucleotide-5'-triphosphate binding aptamers activating RIG-I.


Assuntos
Proteína DEAD-box 58/genética , RNA Helicases/genética , RNA Ribossômico/genética , RNA/genética , Guanosina Trifosfato/genética , Humanos , Ligantes , Fosfatos/metabolismo , RNA/química , RNA Helicases/metabolismo , Receptores Imunológicos , Ribonucleases/genética
4.
RNA Biol ; 18(5): 604-618, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33622174

RESUMO

A persisting obstacle in human immunology is that blood-derived leukocytes are notoriously difficult to manipulate at the RNA level. Therefore, our knowledge about immune-regulatory RNA-networks is largely based on tumour cell-line and rodent knockout models, which do not fully mimic human leukocyte biology. Here, we exploit straightforward cell penetrating peptide (CPP) chemistry to enable efficient loss-of-function phenotyping of regulatory RNAs in primary human blood-derived cells. The classical CPP octaarginine (R8) enabled antisense peptide-nucleic-acid (PNA) oligomer delivery into nearly 100% of human blood-derived macrophages without apparent cytotoxicity even up to micromolar concentrations. In a proof-of-principle experiment, we successfully de-repressed the global microRNA-155 regulome in primary human macrophages using a PNA-R8 oligomer, which phenocopies a CRISPR-Cas9 induced gene knockout. Interestingly, although it is often believed that fairly high concentrations (µM) are needed to achieve antisense activity, our PNA-R8 was effective at 200 nM. RNA-seq characterized microRNA-155 as a broad-acting riboregulator, feedback restraining a late myeloid differentiation-induced pro-inflammatory network, comprising MyD88-signalling and ubiquitin-proteasome components. Our results highlight the important role of the microRNA machinery in fine-control of blood-derived human phagocyte immunity and open the door for further studies on regulatory RNAs in difficult-to-transfect primary human immune cells.


Assuntos
Inflamação/genética , MicroRNAs/fisiologia , Oligonucleotídeos Antissenso/farmacologia , Fagócitos/efeitos dos fármacos , Células Cultivadas , Técnicas de Silenciamento de Genes , Humanos , Inflamação/metabolismo , MicroRNAs/genética , Células Mieloides/efeitos dos fármacos , Células Mieloides/metabolismo , Células Mieloides/fisiologia , Fagócitos/imunologia , Fagócitos/metabolismo , Cultura Primária de Células , Interferência de RNA/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Células U937
5.
J Virol ; 93(21)2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31391268

RESUMO

Cleavage of influenza virus hemagglutinin (HA) by host cell proteases is essential for virus infectivity and spread. We previously demonstrated in vitro that the transmembrane protease TMPRSS2 cleaves influenza A virus (IAV) and influenza B virus (IBV) HA possessing a monobasic cleavage site. Subsequent studies revealed that TMPRSS2 is crucial for the activation and pathogenesis of H1N1pdm and H7N9 IAV in mice. In contrast, activation of H3N2 IAV and IBV was found to be independent of TMPRSS2 expression and supported by an as-yet-undetermined protease(s). Here, we investigated the role of TMPRSS2 in proteolytic activation of IAV and IBV in three human airway cell culture systems: primary human bronchial epithelial cells (HBEC), primary type II alveolar epithelial cells (AECII), and Calu-3 cells. Knockdown of TMPRSS2 expression was performed using a previously described antisense peptide-conjugated phosphorodiamidate morpholino oligomer, T-ex5, that interferes with splicing of TMPRSS2 pre-mRNA, resulting in the expression of enzymatically inactive TMPRSS2. T-ex5 treatment produced efficient knockdown of active TMPRSS2 in all three airway cell culture models and prevented proteolytic activation and multiplication of H7N9 IAV in Calu-3 cells and H1N1pdm, H7N9, and H3N2 IAV in HBEC and AECII. T-ex5 treatment also inhibited the activation and spread of IBV in AECII but did not affect IBV activation in HBEC and Calu-3 cells. This study identifies TMPRSS2 as the major HA-activating protease of IAV in human airway cells and IBV in type II pneumocytes and as a potential target for the development of novel drugs to treat influenza infections.IMPORTANCE Influenza A viruses (IAV) and influenza B viruses (IBV) cause significant morbidity and mortality during seasonal outbreaks. Cleavage of the viral surface glycoprotein hemagglutinin (HA) by host proteases is a prerequisite for membrane fusion and essential for virus infectivity. Inhibition of relevant proteases provides a promising therapeutic approach that may avoid the development of drug resistance. HA of most influenza viruses is cleaved at a monobasic cleavage site, and a number of proteases have been shown to cleave HA in vitro This study demonstrates that the transmembrane protease TMPRSS2 is the major HA-activating protease of IAV in primary human bronchial cells and of both IAV and IBV in primary human type II pneumocytes. It further reveals that human and murine airway cells can differ in their HA-cleaving protease repertoires. Our data will help drive the development of potent and selective protease inhibitors as novel drugs for influenza treatment.


Assuntos
Vírus da Influenza A/fisiologia , Vírus da Influenza B/fisiologia , Influenza Humana/virologia , Serina Endopeptidases/metabolismo , Animais , Brônquios/citologia , Células Cultivadas , Células Epiteliais/virologia , Técnicas de Silenciamento de Genes , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Influenza Humana/enzimologia , Influenza Humana/metabolismo , Camundongos , Infecções por Orthomyxoviridae/enzimologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Alvéolos Pulmonares/citologia , Serina Endopeptidases/genética , Regulação para Cima , Replicação Viral
6.
J Cell Mol Med ; 22(2): 982-998, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29210175

RESUMO

Infectious agents such as lipopolysaccharides (LPS) challenge the functional properties of the alveolar-capillary barrier (ACB) in the lung. In this study, we analyse the site-specific effects of LPS on the ACB and reveal the effects on the individual cell types and the ACB as a functional unit. Monocultures of H441 epithelial cells and co-cultures of H441 with endothelial cells cultured on Transwells® were treated with LPS from the apical or basolateral compartment. Barrier properties were analysed by the transepithelial electrical resistance (TEER), by transport assays, and immunostaining and assessment of tight junctional molecules at protein level. Furthermore, pro-inflammatory cytokines and immune-modulatory molecules were evaluated by ELISA and semiquantitative real-time PCR. Liquid chromatography-mass spectrometry-based proteomics (LS-MS) was used to identify proteins and effector molecules secreted by endothelial cells in response to LPS. In co-cultures treated with LPS from the basolateral compartment, we noticed a significant reduction of TEER, increased permeability and induction of pro-inflammatory cytokines. Conversely, apical treatment did not affect the barrier. No changes were noticed in H441 monoculture upon LPS treatment. However, LPS resulted in an increased expression of pro-inflammatory cytokines such as IL-6 in OEC and in turn induced the reduction of TEER and an increase in SP-A expression in H441 monoculture, and H441/OEC co-cultures after LPS treatment from basolateral compartment. LS-MS-based proteomics revealed factors associated with LPS-mediated lung injury such as ICAM-1, VCAM-1, Angiopoietin 2, complement factors and cathepsin S, emphasizing the role of epithelial-endothelial crosstalk in the ACB in ALI/ARDS.


Assuntos
Capilares/fisiopatologia , Células Endoteliais/patologia , Lipopolissacarídeos/toxicidade , Alvéolos Pulmonares/fisiopatologia , Adulto , Capilares/efeitos dos fármacos , Caveolina 1/metabolismo , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Técnicas de Cocultura , Impedância Elétrica , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/patologia , Fosforilação/efeitos dos fármacos , Proteômica , Alvéolos Pulmonares/efeitos dos fármacos , Proteínas Associadas a Surfactantes Pulmonares/metabolismo , Proteínas de Junções Íntimas/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo
7.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L32-L53, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28860142

RESUMO

The biological and immune-protective properties of surfactant-derived phospholipids and phospholipid subfractions in the context of neonatal inflammatory lung disease are widely unknown. Using a porcine neonatal triple-hit acute respiratory distress syndrome (ARDS) model (repeated airway lavage, overventilation, and LPS instillation into airways), we assessed whether the supplementation of surfactant (S; poractant alfa) with inositol derivatives [inositol 1,2,6-trisphosphate (IP3) or phosphatidylinositol 3,5-bisphosphate (PIP2)] or phosphatidylglycerol subfractions [16:0/18:1-palmitoyloleoyl-phosphatidylglycerol (POPG) or 18:1/18:1-dioleoyl-phosphatidylglycerol (DOPG)] would result in improved clinical parameters and sought to characterize changes in key inflammatory pathways behind these improvements. Within 72 h of mechanical ventilation, the oxygenation index (S+IP3, S+PIP2, and S+POPG), the ventilation efficiency index (S+IP3 and S+POPG), the compliance (S+IP3 and S+POPG) and resistance (S+POPG) of the respiratory system, and the extravascular lung water index (S+IP3 and S+POPG) significantly improved compared with S treatment alone. The inositol derivatives (mainly S+IP3) exerted their actions by suppressing acid sphingomyelinase activity and dependent ceramide production, linked with the suppression of the inflammasome nucleotide-binding domain, leucine-rich repeat-containing protein-3 (NLRP3)-apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC)-caspase-1 complex, and the profibrotic response represented by the cytokines transforming growth factor-ß1 and IFN-γ, matrix metalloproteinase (MMP)-1/8, and elastin. In addition, IκB kinase activity was significantly reduced. S+POPG and S+DOPG treatment inhibited polymorphonuclear leukocyte activity (MMP-8 and myeloperoxidase) and the production of interleukin-6, maintained alveolar-capillary barrier functions, and reduced alveolar epithelial cell apoptosis, all of which resulted in reduced pulmonary edema. S+DOPG also limited the profibrotic response. We conclude that highly concentrated inositol derivatives and phosphatidylglycerol subfractions in surfactant preparations mitigate key inflammatory pathways in inflammatory lung disease and that their clinical application may be of interest for future treatment of the acute exudative phase of neonatal ARDS.


Assuntos
Modelos Animais de Doenças , Inositol/farmacologia , Fosfatidilgliceróis/farmacologia , Edema Pulmonar/tratamento farmacológico , Surfactantes Pulmonares/farmacologia , Síndrome do Desconforto Respiratório do Recém-Nascido/tratamento farmacológico , Animais , Animais Recém-Nascidos , Apoptose , Líquido da Lavagem Broncoalveolar , Citocinas/genética , Citocinas/metabolismo , Feminino , Humanos , Masculino , NF-kappa B/genética , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Edema Pulmonar/metabolismo , Edema Pulmonar/patologia , Troca Gasosa Pulmonar , Distribuição Aleatória , Respiração Artificial , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/patologia , Suínos , Pesquisa Translacional Biomédica , Complexo Vitamínico B/farmacologia
8.
Angew Chem Int Ed Engl ; 57(36): 11564-11568, 2018 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-29972713

RESUMO

Transgene expression of green fluorescent protein (GFP) has facilitated the spatiotemporal investigation of host-pathogen interactions; however, introduction of the GFP gene remains challenging in drug-resistant bacteria. Herein, we report a novel far-red fluorescent nucleic acid stain, 6-TramTO-3, which efficiently labels bacteria through a DNA binding mode without affecting growth and viability. Exemplarily, we stained Klebsiella pneumoniae, a major threat to hospitalized patients, and deciphered divergent interaction strategies of antibiotic-resistant and antibiotic-sensitive Klebsiella strains with immune cells. 6-TramTO-3 constitutes an off-the-shelf reagent for real-time analysis of bacterial infection, including strains for which the use of genetically encoded reporters is not feasible. Eventually, our approach may aid the development of strategies to combat a major worldwide health threat: multidrug-resistant bacteria.


Assuntos
Carbocianinas/análise , DNA Bacteriano/análise , Corantes Fluorescentes/análise , Infecções por Klebsiella/microbiologia , Klebsiella pneumoniae/citologia , Farmacorresistência Bacteriana Múltipla , Humanos , Klebsiella pneumoniae/isolamento & purificação , Macrófagos/microbiologia , Espectrometria de Fluorescência , Coloração e Rotulagem
9.
mBio ; 11(1)2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32071273

RESUMO

A major obstacle in infection biology is the limited ability to recapitulate human disease trajectories in traditional cell culture and animal models, which impedes the translation of basic research into clinics. Here, we introduce a three-dimensional (3D) intestinal tissue model to study human enteric infections at a level of detail that is not achieved by conventional two-dimensional monocultures. Our model comprises epithelial and endothelial layers, a primary intestinal collagen scaffold, and immune cells. Upon Salmonella infection, the model mimics human gastroenteritis, in that it restricts the pathogen to the epithelial compartment, an advantage over existing mouse models. Application of dual transcriptome sequencing to the Salmonella-infected model revealed the communication of epithelial, endothelial, monocytic, and natural killer cells among each other and with the pathogen. Our results suggest that Salmonella uses its type III secretion systems to manipulate STAT3-dependent inflammatory responses locally in the epithelium without accompanying alterations in the endothelial compartment. Our approach promises to reveal further human-specific infection strategies employed by Salmonella and other pathogens.IMPORTANCE Infection research routinely employs in vitro cell cultures or in vivo mouse models as surrogates of human hosts. Differences between murine and human immunity and the low level of complexity of traditional cell cultures, however, highlight the demand for alternative models that combine the in vivo-like properties of the human system with straightforward experimental perturbation. Here, we introduce a 3D tissue model comprising multiple cell types of the human intestinal barrier, a primary site of pathogen attack. During infection with the foodborne pathogen Salmonella enterica serovar Typhimurium, our model recapitulates human disease aspects, including pathogen restriction to the epithelial compartment, thereby deviating from the systemic infection in mice. Combination of our model with state-of-the-art genetics revealed Salmonella-mediated local manipulations of human immune responses, likely contributing to the establishment of the pathogen's infection niche. We propose the adoption of similar 3D tissue models to infection biology, to advance our understanding of molecular infection strategies employed by bacterial pathogens in their human host.


Assuntos
Técnicas de Cocultura/métodos , Interações Hospedeiro-Patógeno/fisiologia , Intestinos/microbiologia , Infecções por Salmonella/microbiologia , Animais , Sistemas CRISPR-Cas , Células CACO-2 , Modelos Animais de Doenças , Células Epiteliais/microbiologia , Epitélio/microbiologia , Gastroenterite/microbiologia , Regulação Bacteriana da Expressão Gênica , Humanos , Células Matadoras Naturais , Camundongos , Fator de Transcrição STAT3/metabolismo , Infecções por Salmonella/imunologia , Salmonella typhimurium/genética , Salmonella typhimurium/imunologia , Transcriptoma , Sistemas de Secreção Tipo III
10.
Int J Pharm ; 591: 119993, 2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-33086089

RESUMO

P-glycoprotein (P-gp) associated multidrug resistance (MDR) represents a major failure in cancer treatment. The overexpression of P-gp is responsible for ATP-dependent efflux of drugs that decrease their intracellular accumulation. An effective downregulation of MDR1 gene using small interfering RNA (siRNA) is one of the safe and effective tools to overcome the P-gp triggered MDR. Therefore, the development of an efficient and non-toxic carrier system for siRNA delivery is a fundamental challenge for effective cancer treatment. Polyamidoamine (PAMAM) dendrimer has been used for efficient delivery of siRNA (dendriplexes) to the tumor cells but the associated toxicity problems render its use in biological applications. A non-covalent lipid modification (lipodendriplexes) is supposed to offer a promising strategy to overcome the demerits linked to the naked dendriplexes system. In the current study, we deliver siRNA, designed against MDR1 gene (si-MDR1), in colorectal carcinoma cells (Caco-2), having overexpression of P-gp, to check the role of MDR1 gene in tumor progression and multidrug resistance using two dimensional (2D) and three dimensional (3D) environment. Imatinib mesylate (IM), a P-gp substrate, was used as model drug. Our results revealed that the effective knockdown by lipodendriplexes system can significantly reduce the tumor cell migration in 2D (p < 0.001) and 3D (p < 0.001) cell cultures as compared to unmodified dendriplexes and si-Control groups. It was also observed that lipodendriplexes aided downregulation of MDR1 gene effectively, re-sensitized the Caco-2 cells for IM uptake and showed a significantly (p < 0.001) higher apoptosis. Our findings imply that our lipodendriplexes system has a great potential for siRNA delivery, however, further in vivo application using a suitable targeted system can play a major role for better cancer therapeutics.


Assuntos
Apoptose , Resistencia a Medicamentos Antineoplásicos , Células CACO-2 , Linhagem Celular Tumoral , Regulação para Baixo , Humanos , Inibidores de Proteínas Quinases , RNA Interferente Pequeno
11.
PLoS One ; 13(2): e0193066, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29451908

RESUMO

CRISPR/Cas9-based approaches have greatly facilitated targeted genomic deletions. Contrary to coding genes however, which can be functionally knocked out by frame-shift mutagenesis, non-coding RNA (ncRNA) gene knockouts have remained challenging. Here we present a universal ncRNA knockout approach guided by epigenetic hallmarks, which enables robust gene silencing even in provisionally annotated gene loci. We build on previous work reporting the presence of overlapping histone H3 lysine 4 tri-methylation (H3K4me3) and DNaseI hypersensitivity sites around the transcriptional start sites of most genes. We demonstrate that excision of this gene-proximal signature leads to loss of microRNA and lincRNA transcription and reveals ncRNA phenotypes. Exemplarily we demonstrate silencing of the constitutively transcribed MALAT1 lincRNA gene as well as of the inducible miR-146a and miR-155 genes in human monocytes. Our results validate a role of miR-146a and miR-155 in negative feedback control of the activity of inflammation master-regulator NFκB and suggest that cell-cycle control is a unique feature of miR-155. We suggest that our epigenetically guided CRISPR approach may improve existing ncRNA knockout strategies and contribute to the development of high-confidence ncRNA phenotyping applications.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Desoxirribonuclease I/metabolismo , Técnicas de Inativação de Genes/métodos , Inativação Gênica , MicroRNAs/genética , Northern Blotting , Desoxirribonuclease I/genética , Citometria de Fluxo , Regulação da Expressão Gênica/genética , Histonas/genética , Humanos , Monócitos/metabolismo , RNA Longo não Codificante/genética , Reação em Cadeia da Polimerase em Tempo Real
12.
PLoS One ; 13(9): e0203466, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30212482

RESUMO

Despite the important functions of PPARγ in various cell types of the lung, PPARγ-deficiency in club cells induces only mild emphysema. Peroxisomes are distributed in a similar way as PPARγ in the lung and are mainly enriched in club and AECII cells. To date, the effects of PPARγ-deficiency on the overall peroxisomal compartment and its metabolic alterations in pulmonary club cells are unknown. Therefore, we characterized wild-type and club cell-specific PPARγ knockout-mice lungs and used C22 cells to investigate the peroxisomal compartment and its metabolic roles in the distal airway epithelium by means of 1) double-immunofluorescence labelling for peroxisomal proteins, 2) laser-assisted microdissection of the bronchiolar epithelium and subsequent qRT-PCR, 3) siRNA-transfection of PPARγand PPRE dual-luciferase reporter activity in C22 cells, 4) PPARg inhibition by GW9662, 5) GC-MS based lipid analysis. Our results reveal elevated levels of fatty acids, increased expression of PPARα and PPRE activity, a strong overall upregulation of the peroxisomal compartment and its associated gene expression (biogenesis, α-oxidation, ß-oxidation, and plasmalogens) in PPARγ-deficient club cells. Interestingly, catalase was significantly increased and mistargeted into the cytoplasm, suggestive for oxidative stress by the PPARγ-deficiency in club cells. Taken together, PPARα-mediated metabolic induction and proliferation of peroxisomes via a PPRE-dependent mechanism could compensate PPARγ-deficiency in club cells.


Assuntos
Brônquios/metabolismo , Regulação da Expressão Gênica , PPAR alfa/biossíntese , PPAR gama/deficiência , Peroxissomos/metabolismo , Enfisema Pulmonar/metabolismo , Anilidas/farmacologia , Animais , Brônquios/patologia , Camundongos , Camundongos Knockout , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , PPAR alfa/antagonistas & inibidores , PPAR alfa/genética , Peroxissomos/genética , Enfisema Pulmonar/genética , Enfisema Pulmonar/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA