Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 5583, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961085

ABSTRACT

The function of many bacterial processes depends on the formation of functional membrane microdomains (FMMs), which resemble the lipid rafts of eukaryotic cells. However, the mechanism and the biological function of these membrane microdomains remain unclear. Here, we show that FMMs in the pathogen methicillin-resistant Staphylococcus aureus (MRSA) are dedicated to confining and stabilizing proteins unfolded due to cellular stress. The FMM scaffold protein flotillin forms a clamp-shaped oligomer that holds unfolded proteins, stabilizing them and favoring their correct folding. This process does not impose a direct energy cost on the cell and is crucial to survival of ATP-depleted bacteria, and thus to pathogenesis. Consequently, FMM disassembling causes the accumulation of unfolded proteins, which compromise MRSA viability during infection and cause penicillin re-sensitization due to PBP2a unfolding. Thus, our results indicate that FMMs mediate ATP-independent stabilization of unfolded proteins, which is essential for bacterial viability during infection.


Subject(s)
Bacterial Proteins , Membrane Microdomains , Membrane Proteins , Methicillin-Resistant Staphylococcus aureus , Membrane Proteins/metabolism , Membrane Microdomains/metabolism , Methicillin-Resistant Staphylococcus aureus/metabolism , Bacterial Proteins/metabolism , Protein Unfolding , Adenosine Triphosphate/metabolism , Penicillin-Binding Proteins/metabolism , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/chemistry , Humans , Protein Stability , Staphylococcal Infections/microbiology , Staphylococcal Infections/metabolism , Animals , Mice
2.
Dalton Trans ; 52(41): 14762-14773, 2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37548588

ABSTRACT

A new series of Zn(II) and Cu(II)-based porphyrin complexes 5a and 5b doubly functionalised with carbazole units were developed to be used as hole-transporting materials (HTMs) in perovskite solar cells (PSCs). These complexes were obtained via a nucleophilic substitution reaction mediated by PhI(OAc)2/NaAuCl4·2H2O, or using C-N transition metal-assisted coupling. The hole extraction capability of 5a and 5b was assessed using cyclic voltammetry; this study confirmed the better alignment of the Zn(II) complex 5a with the perovskite valence band level, compared to the Cu(II) complex 5b. The optimised geometry and molecular orbitals of both complexes also corroborate the higher potential of 5a as a HTM. Photoluminescence characterisation showed that the presence of 5a and 5b as HTMs on the perovskite surface resulted in the quenching of the emission, matching the hole transfer phenomenon. The photovoltaic performance was evaluated and compared with those of reference cells made with the standard HTM spiro-OMeTAD. The optimised 5-based devices showed improvements in all photovoltaic characteristics; their open circuit voltage (Voc) reached close to 1 V and short-circuit current density (Jsc) values were 13.79 and 9.14 mA cm-2 for 5a and 5b, respectively, disclosing the effect of the metallic centre. A maximum power conversion efficiency (PCE) of 10.01% was attained for 5a, which is 65% of the PCE generated by using the spiro-OMeTAD reference. This study demonstrates that C-N linked donor-type porphyrin derivatives are promising novel HTMs for developing efficient and reproducible PSCs.

3.
Nat Commun ; 13(1): 7174, 2022 11 22.
Article in English | MEDLINE | ID: mdl-36418309

ABSTRACT

Staphylococcus aureus is increasingly recognized as a facultative intracellular pathogen, although the significance and pervasiveness of its intracellular lifestyle remain controversial. Here, we applied fluorescence microscopy-based infection assays and automated image analysis to profile the interaction of 191 S. aureus isolates from patients with bone/joint infections, bacteremia, and infective endocarditis, with four host cell types, at five times post-infection. This multiparametric analysis revealed that almost all isolates are internalized and that a large fraction replicate and persist within host cells, presenting distinct infection profiles in non-professional vs. professional phagocytes. Phenotypic clustering highlighted interesting sub-groups, including one comprising isolates exhibiting high intracellular replication and inducing delayed host death in vitro and in vivo. These isolates are deficient for the cysteine protease staphopain A. This study establishes S. aureus intracellular lifestyle as a prevalent feature of infection, with potential implications for the effective treatment of staphylococcal infections.


Subject(s)
Bacteremia , Staphylococcal Infections , Humans , Staphylococcus aureus , Microscopy , Life Style
4.
Nat Commun ; 13(1): 1525, 2022 03 21.
Article in English | MEDLINE | ID: mdl-35314690

ABSTRACT

A central question concerning natural competence is why orthologs of competence genes are conserved in non-competent bacterial species, suggesting they have a role other than in transformation. Here we show that competence induction in the human pathogen Staphylococcus aureus occurs in response to ROS and host defenses that compromise bacterial respiration during infection. Bacteria cope with reduced respiration by obtaining energy through fermentation instead. Since fermentation is energetically less efficient than respiration, the energy supply must be assured by increasing the glycolytic flux. The induction of natural competence increases the rate of glycolysis in bacteria that are unable to respire via upregulation of DNA- and glucose-uptake systems. A competent-defective mutant showed no such increase in glycolysis, which negatively affects its survival in both mouse and Galleria infection models. Natural competence foster genetic variability and provides S. aureus with additional nutritional and metabolic possibilities, allowing it to proliferate during infection.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carbohydrate Metabolism , Fermentation , Glycolysis/genetics , Mice , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism
5.
Autophagy ; 18(8): 1785-1800, 2022 08.
Article in English | MEDLINE | ID: mdl-34781820

ABSTRACT

Modulation of the host cell cycle has emerged as a common theme among the pathways regulated by bacterial pathogens, arguably to promote host cell colonization. However, in most cases the exact benefit ensuing from such interference to the infection process remains unclear. Previously, we have shown that Salmonella actively induces G2/M arrest of host cells, and that infection is severely inhibited in cells arrested in G1. In this study, we demonstrate that Salmonella vacuolar replication is inhibited in host cells blocked in G1, whereas the cytosolic replication of the closely related pathogen Shigella is not affected. Mechanistically, we show that cells arrested in G1, but not cells arrested in G2, present dysregulated endolysosomal trafficking, displaying an abnormal accumulation of vesicles positive for late endosomal and lysosomal markers. In addition, the macroautophagic/autophagic flux and degradative lysosomal function are strongly impaired. This endolysosomal trafficking dysregulation results in sustained activation of the SPI-1 type III secretion system and lack of vacuole repair by the autophagy pathway, ultimately compromising the maturation and integrity of the Salmonella-containing vacuole. As such, Salmonella is released in the host cytosol. Collectively, our findings demonstrate that the modulation of the host cell cycle occurring during Salmonella infection is related to a disparity in the permissivity of cells arrested in G1 and G2/M, due to their intrinsic characteristics.Abbreviations: CDK4: cyclin dependent kinase 4; CDK6: cyclin dependent kinase 6; CDK4-CDK6i: CDK4-CDK6 inhibitor IV; cfu: colony-forming units; CHQ: chloroquine; DMSO: dimethyl sulfoxide; EEA1: early endosome antigen 1; FITC: fluorescein isothiocyanate; GFP: green fluorescent protein; hpi: hours post-infection; LAMP1: lysosomal associated membrane protein 1; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MOI: multiplicity of infection; RAB7: RAB7, member RAS oncogene family; SCV: Salmonella-containing vacuole; SPI-1: Salmonella pathogenicity island-1; SPI-2: Salmonella pathogenicity island-2; TFEB: transcription factor EB; T3SS: type III secretion system.


Subject(s)
Type III Secretion Systems , Vacuoles , Autophagy , Bacterial Proteins/metabolism , Cell Cycle , Lysosomes/metabolism , Salmonella/metabolism , Type III Secretion Systems/metabolism , Vacuoles/metabolism
6.
Nat Commun ; 12(1): 3392, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099666

ABSTRACT

Cells infected with pathogens can contribute to clearing infections by releasing signals that instruct neighbouring cells to mount a pro-inflammatory cytokine response, or by other mechanisms that reduce bystander cells' susceptibility to infection. Here, we show the opposite effect: epithelial cells infected with Salmonella Typhimurium secrete host factors that facilitate the infection of bystander cells. We find that the endoplasmic reticulum stress response is activated in both infected and bystander cells, and this leads to activation of JNK pathway, downregulation of transcription factor E2F1, and consequent reprogramming of microRNA expression in a time-dependent manner. These changes are not elicited by infection with other bacterial pathogens, such as Shigella flexneri or Listeria monocytogenes. Remarkably, the protein HMGB1 present in the secretome of Salmonella-infected cells is responsible for the activation of the IRE1 branch of the endoplasmic reticulum stress response in non-infected, neighbouring cells. Furthermore, E2F1 downregulation and the associated microRNA alterations promote Salmonella replication within infected cells and prime bystander cells for more efficient infection.


Subject(s)
Bystander Effect/genetics , E2F1 Transcription Factor/metabolism , MicroRNAs/metabolism , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Animals , Bystander Effect/immunology , Disease Models, Animal , Down-Regulation/immunology , E2F1 Transcription Factor/genetics , Endoplasmic Reticulum Stress/immunology , Endoribonucleases/metabolism , HMGB1 Protein/metabolism , HeLa Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Listeria monocytogenes/immunology , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Protein Serine-Threonine Kinases/metabolism , RNA-Seq , Salmonella Infections/genetics , Salmonella Infections/microbiology , Salmonella typhimurium/pathogenicity , Shigella flexneri/immunology , Swine
7.
Nat Microbiol ; 5(1): 192-205, 2020 01.
Article in English | MEDLINE | ID: mdl-31792428

ABSTRACT

MicroRNAs (miRNAs) are increasingly recognized for their role in infection by bacterial pathogens, although the effect of each individual miRNA remains largely unknown. Here, we used a comparative genome-wide microscopy-based functional screening approach to identify miRNAs controlling infection by two bacterial pathogens-Salmonella enterica serovar Typhimurium and Shigella flexneri. Despite the similarities between these pathogens, we found infections to be controlled by largely non-overlapping subsets of miRNAs, seemingly reflecting different requirements prompted by their distinct intracellular lifestyles. By characterizing a small subset of miRNAs chosen among the strongest inhibitors of Shigella infection, we discovered that miR-3668, miR-4732-5p and miR-6073 exert a selective effect on Shigella infection by impairing bacterial actin-based motility by downregulating N-WASP. Additionally, by identifying let-7i-3p miRNA as a strong inhibitor of Salmonella replication and performing in-depth analysis of its mechanisms of action, we showed that this miRNA specifically inhibits Salmonella infection via modulation of endolysosomal trafficking and the vacuolar environment by targeting the host RGS2 protein. These findings illustrate two paradigms underlying miRNA-mediated regulation of bacterial infection, acting as part of the host response to infection, or as part of bacterial strategies to modulate the host environment and favour pathogenesis.


Subject(s)
Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/microbiology , MicroRNAs/genetics , Salmonella typhimurium/physiology , Shigella flexneri/physiology , Animals , Gene Expression Regulation , Genomics , HeLa Cells , Host-Pathogen Interactions , Humans , MicroRNAs/metabolism , Species Specificity , Swine
8.
Microbiol Spectr ; 7(3)2019 05.
Article in English | MEDLINE | ID: mdl-31152522

ABSTRACT

MicroRNAs (miRNAs) are a well-characterized class of small noncoding RNAs that act as major posttranscriptional regulators of gene expression. Accordingly, miRNAs have been associated with a wide range of fundamental biological processes and implicated in human diseases. During the past decade, miRNAs have also been recognized for their role in the complex interplay between the host and bacterial pathogens, either as part of the host response to counteract infection or as a molecular strategy employed by bacteria to subvert host pathways for their own benefit. Importantly, the characterization of downstream miRNA targets and their underlying mechanisms of action has uncovered novel molecular factors and pathways relevant to infection. In this article, we review the current knowledge of the miRNA response to bacterial infection, focusing on different bacterial pathogens, including Salmonella enterica, Listeria monocytogenes, Mycobacterium spp., and Helicobacter pylori, among others.


Subject(s)
Bacterial Infections/metabolism , Bacterial Physiological Phenomena , Host-Pathogen Interactions/physiology , MicroRNAs/physiology , Animals , Bacteria/pathogenicity , Gene Expression Regulation , Helicobacter pylori , Host-Pathogen Interactions/genetics , Humans , Listeria monocytogenes , MicroRNAs/genetics , Mycobacterium , Salmonella enterica
9.
Methods ; 152: 55-64, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30292796

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs that post-transcriptionally modulate gene expression and orchestrate a wide range of biological and pathological processes. The use of high-throughput screening technologies, in particular microscopy-based screenings (also known as high-content screenings), coupled with genome-wide libraries for modulation of miRNA levels, allow for comprehensive functional analysis of each member of the miRNome in different phenotypic cell-based assays. The wealth of information obtained from such screenings spans across various fields of research, including cancer, cardiovascular, cell reprogramming, and infection biology. Here, we provide an overview of the rationale for performing screenings using synthetic libraries of miRNA mimics and inhibitors, and of the microscopy-based miRNA screenings performed to date. Moreover, a list of resources available for such endeavor is provided. Finally, we describe a detailed procedure for a case study where microscopy-based screening using a library of miRNA mimics was performed to identify miRNAs that control infection of epithelial cells by the bacterial pathogen Salmonella. The methodologies described here can be easily adapted for screenings addressing other biological questions.


Subject(s)
MicroRNAs/physiology , Salmonella Infections/genetics , Epithelial Cells/microbiology , Gene Expression Regulation , Humans , Salmonella , Transfection/methods
10.
Trends Microbiol ; 27(3): 206-218, 2019 03.
Article in English | MEDLINE | ID: mdl-30477908

ABSTRACT

MicroRNAs are a class of small noncoding RNAs that act as major post-transcriptional regulators of gene expression. They are currently recognized for their important role in the intricate interaction between host and bacterial pathogens, either as part of the host immune response to neutralize infection, or as a molecular strategy employed by bacteria to hijack host pathways for their own benefit. Here, we summarize recent advances on the function of miRNAs during infection of mammalian hosts by bacterial pathogens, highlighting key cellular pathways. In addition, we discuss emerging themes in this field, including the participation of miRNAs in host-microbiota crosstalk and cell-to-cell communication.


Subject(s)
Bacteria/genetics , Bacterial Infections/immunology , Gene Expression Regulation , Host-Pathogen Interactions/genetics , MicroRNAs/genetics , Animals , Bacteria/immunology , Bacteria/pathogenicity , Gastrointestinal Microbiome , Host-Pathogen Interactions/immunology , Humans
11.
EMBO J ; 37(23)2018 12 03.
Article in English | MEDLINE | ID: mdl-30389666

ABSTRACT

While mucosal inflammation is a major source of stress during enteropathogen infection, it remains to be fully elucidated how the host benefits from this environment to clear the pathogen. Here, we show that host stress induced by different stimuli mimicking inflammatory conditions strongly reduces the binding of Shigella flexneri to epithelial cells. Mechanistically, stress activates acid sphingomyelinase leading to host membrane remodeling. Consequently, knockdown or pharmacological inhibition of the acid sphingomyelinase blunts the stress-dependent inhibition of Shigella binding to host cells. Interestingly, stress caused by intracellular Shigella replication also results in remodeling of the host cell membrane, in vitro and in vivo, which precludes re-infection by this and other non-motile pathogens. In contrast, Salmonella Typhimurium overcomes the shortage of permissive entry sites by gathering effectively at the remaining platforms through its flagellar motility. Overall, our findings reveal host membrane remodeling as a novel stress-responsive cell-autonomous defense mechanism that protects epithelial cells from infection by non-motile bacterial pathogens.


Subject(s)
Cell Membrane/immunology , Dysentery, Bacillary/immunology , Epithelial Cells/immunology , Immunity, Innate , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Shigella flexneri/immunology , Stress, Physiological/immunology , Animals , Cell Membrane/pathology , Dysentery, Bacillary/pathology , Epithelial Cells/pathology , Guinea Pigs , Salmonella Infections/pathology
12.
Sci Rep ; 7(1): 9328, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28839189

ABSTRACT

Many pathogenic bacteria utilize specialized secretion systems to deliver proteins called effectors into eukaryotic cells for manipulation of host pathways. The vast majority of known effector targets are host proteins, whereas a potential targeting of host nucleic acids remains little explored. There is only one family of effectors known to target DNA directly, and effectors binding host RNA are unknown. Here, we take a two-pronged approach to search for RNA-binding effectors, combining biocomputational prediction of RNA-binding domains (RBDs) in a newly assembled comprehensive dataset of bacterial secreted proteins, and experimental screening for RNA binding in mammalian cells. Only a small subset of effectors were predicted to carry an RBD, indicating that if RNA targeting was common, it would likely involve new types of RBDs. Our experimental evaluation of effectors with predicted RBDs further argues for a general paucity of RNA binding activities amongst bacterial effectors. We obtained evidence that PipB2 and Lpg2844, effector proteins of Salmonella and Legionella species, respectively, may harbor novel biochemical activities. Our study presenting the first systematic evaluation of the RNA-targeting potential of bacterial effectors offers a basis for discussion of whether or not host RNA is a prominent target of secreted bacterial proteins.


Subject(s)
Bacterial Proteins/metabolism , Eukaryotic Cells/metabolism , Legionella/metabolism , RNA-Binding Proteins/metabolism , RNA/metabolism , Salmonella/metabolism , Virulence Factors/metabolism , Bacterial Proteins/genetics , Binding Sites , Computational Biology , Data Mining , Legionella/genetics , RNA-Binding Proteins/genetics , Salmonella/genetics , Virulence Factors/genetics
13.
PLoS Pathog ; 13(4): e1006327, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28394930

ABSTRACT

MicroRNAs play an important role in the interplay between bacterial pathogens and host cells, participating as host defense mechanisms, as well as exploited by bacteria to subvert host cellular functions. Here, we show that microRNAs modulate infection by Shigella flexneri, a major causative agent of bacillary dysentery in humans. Specifically, we characterize the dual regulatory role of miR-29b-2-5p during infection, showing that this microRNA strongly favors Shigella infection by promoting both bacterial binding to host cells and intracellular replication. Using a combination of transcriptome analysis and targeted high-content RNAi screening, we identify UNC5C as a direct target of miR-29b-2-5p and show its pivotal role in the modulation of Shigella binding to host cells. MiR-29b-2-5p, through repression of UNC5C, strongly enhances filopodia formation thus increasing Shigella capture and promoting bacterial invasion. The increase of filopodia formation mediated by miR-29b-2-5p is dependent on RhoF and Cdc42 Rho-GTPases. Interestingly, the levels of miR-29b-2-5p, but not of other mature microRNAs from the same precursor, are decreased upon Shigella replication at late times post-infection, through degradation of the mature microRNA by the exonuclease PNPT1. While the relatively high basal levels of miR-29b-2-5p at the start of infection ensure efficient Shigella capture by host cell filopodia, dampening of miR-29b-2-5p levels later during infection may constitute a bacterial strategy to favor a balanced intracellular replication to avoid premature cell death and favor dissemination to neighboring cells, or alternatively, part of the host response to counteract Shigella infection. Overall, these findings reveal a previously unappreciated role of microRNAs, and in particular miR-29b-2-5p, in the interaction of Shigella with host cells.


Subject(s)
Host-Pathogen Interactions/genetics , MicroRNAs/genetics , Shigella/genetics , Shigella/virology , Virus Replication/genetics , Cell Line , DNA Replication/genetics , Gene Expression Profiling/methods , Host-Pathogen Interactions/immunology , Humans , Pseudopodia/immunology , RNA Interference/physiology
14.
Nucleic Acids Res ; 45(11): e96, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28334975

ABSTRACT

RNA-binding proteins (RBPs) have been established as core components of several post-transcriptional gene regulation mechanisms. Experimental techniques such as cross-linking and co-immunoprecipitation have enabled the identification of RBPs, RNA-binding domains (RBDs) and their regulatory roles in the eukaryotic species such as human and yeast in large-scale. In contrast, our knowledge of the number and potential diversity of RBPs in bacteria is poorer due to the technical challenges associated with the existing global screening approaches. We introduce APRICOT, a computational pipeline for the sequence-based identification and characterization of proteins using RBDs known from experimental studies. The pipeline identifies functional motifs in protein sequences using position-specific scoring matrices and Hidden Markov Models of the functional domains and statistically scores them based on a series of sequence-based features. Subsequently, APRICOT identifies putative RBPs and characterizes them by several biological properties. Here we demonstrate the application and adaptability of the pipeline on large-scale protein sets, including the bacterial proteome of Escherichia coli. APRICOT showed better performance on various datasets compared to other existing tools for the sequence-based prediction of RBPs by achieving an average sensitivity and specificity of 0.90 and 0.91 respectively. The command-line tool and its documentation are available at https://pypi.python.org/pypi/bio-apricot.


Subject(s)
RNA-Binding Proteins/chemistry , Sequence Analysis, Protein , Software , Binding Sites , Computational Biology , Escherichia coli Proteins/chemistry , Molecular Sequence Annotation , Protein Domains
15.
J Cell Biol ; 216(4): 1071-1089, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28330939

ABSTRACT

Obligate intracellular bacteria such as Chlamydia trachomatis depend on metabolites of the host cell and thus protect their sole replication niche by interfering with the host cells' stress response. Here, we investigated the involvement of host microRNAs (miRNAs) in maintaining the viability of C. trachomatis-infected primary human cells. We identified miR-30c-5p as a prominently up-regulated miRNA required for the stable down-regulation of p53, a major suppressor of metabolite supply in C. trachomatis-infected cells. Loss of miR-30c-5p led to the up-regulation of Drp1, a mitochondrial fission regulator and a target gene of p53, which, in turn, severely affected chlamydial growth and had a marked effect on the mitochondrial network. Drp1-induced mitochondrial fragmentation prevented replication of C. trachomatis even in p53-deficient cells. Additionally, Chlamydia maintain mitochondrial integrity during reactive oxygen species-induced stress that occurs naturally during infection. We show that C. trachomatis require mitochondrial ATP for normal development and hence postulate that they preserve mitochondrial integrity through a miR-30c-5p-dependent inhibition of Drp1-mediated mitochondrial fission.


Subject(s)
Chlamydia Infections/microbiology , Chlamydia trachomatis/genetics , DNA Replication/genetics , MicroRNAs/genetics , Mitochondria/microbiology , Mitochondrial Dynamics/genetics , Cell Line, Tumor , Cells, Cultured , Down-Regulation/genetics , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Up-Regulation/genetics
16.
J Biomol Screen ; 20(8): 1003-17, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25824005

ABSTRACT

MicroRNAs (miRNAs) are a class of genome-encoded small RNAs that post-transcriptionally regulate gene expression by repressing target transcripts containing partially or fully complementary binding sites.Despite their relatively low number, miRNAs have been shown to directly regulate a large fraction of the transcriptome. In agreement with their pervasive role in the regulation of eukaryotic gene expression, miRNAs have been implicated in virtually all biological processes, including different pathologies.The use of screening technologies to systematically analyze miRNA function in cell-based assays offers a unique opportunity to gain new insights into complex biological and disease-relevant processes. Given the low complexity of the miRNome and the similarities to small interfering RNA (siRNA) screening experimental approaches, phenotypic screening using genome-wide libraries of miRNA mimics or inhibitors is not, per se, technically challenging. The identification of miRNA targets and, more importantly, the characterization of their mechanisms of action through the identification of the key targets underlying observed phenotypes remain the major challenges of this approach.This article provides an overview of cell-based screenings for miRNA function that were performed in different biological contexts. The advantages and limitations of computational and experimental approaches commonly used to identify miRNA targets are also discussed.


Subject(s)
High-Throughput Screening Assays , MicroRNAs/genetics , RNA Interference , RNA, Messenger/genetics , Animals , Cell Line , Computational Biology/methods , Genomics/methods , Humans , Reproducibility of Results
17.
Nat Commun ; 5: 4718, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-25146723

ABSTRACT

Increasing evidence suggests an important role for miRNAs in the molecular interplay between bacterial pathogens and host cells. Here we perform a fluorescence microscopy-based screen using a library of miRNA mimics and demonstrate that miRNAs modulate Salmonella infection. Several members of the miR-15 miRNA family were among the 17 miRNAs that more efficiently inhibit Salmonella infection. We discovered that these miRNAs are downregulated during Salmonella infection, through the inhibition of the transcription factor E2F1. Analysis of miR-15 family targets revealed that derepression of cyclin D1 and the consequent promotion of G1/S transition are crucial for Salmonella intracellular proliferation. In addition, Salmonella induces G2/M cell cycle arrest in infected cells, further promoting its replication. Overall, these findings uncover a mechanism whereby Salmonella renders host cells more susceptible to infection by controlling cell cycle progression through the active modulation of host cell miRNAs.


Subject(s)
Host-Pathogen Interactions/genetics , MicroRNAs/genetics , Salmonella Infections/genetics , Animals , Cell Cycle Checkpoints , Cyclin D1/genetics , E2F1 Transcription Factor/genetics , Gene Expression Regulation , HeLa Cells/drug effects , HeLa Cells/microbiology , High-Throughput Nucleotide Sequencing/methods , Humans , Lipopolysaccharides/pharmacology , Mice , Multigene Family , RAW 264.7 Cells/microbiology , Salmonella typhimurium/genetics , Salmonella typhimurium/pathogenicity
18.
FEBS Lett ; 588(22): 4140-7, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25128459

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs with a central role in the post-transcriptional control of gene expression, that have been implicated in a wide-range of biological processes. Regulation of miRNA expression is increasingly recognized as a crucial part of the host response to infection by bacterial pathogens, as well as a novel molecular strategy exploited by bacteria to manipulate host cell pathways. Here, we review the current knowledge of bacterial pathogens that modulate host miRNA expression, focusing on mammalian host cells, and the implications of miRNA regulation on the outcome of infection. The emerging role of commensal bacteria, as part of the gut microbiota, on host miRNA expression in the presence or absence of bacterial pathogens is also discussed.


Subject(s)
Bacterial Physiological Phenomena , Host-Pathogen Interactions/genetics , MicroRNAs/genetics , Animals , Gastrointestinal Tract/microbiology , Gene Expression Regulation , Humans
19.
Nucleic Acids Res ; 41(2): 978-94, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23172285

ABSTRACT

Animal miRNAs silence the expression of mRNA targets through translational repression, deadenylation and subsequent mRNA degradation. Silencing requires association of miRNAs with an Argonaute protein and a GW182 family protein. In turn, GW182 proteins interact with poly(A)-binding protein (PABP) and the PAN2-PAN3 and CCR4-NOT deadenylase complexes. These interactions are required for the deadenylation and decay of miRNA targets. Recent studies have indicated that miRNAs repress translation before inducing target deadenylation and decay; however, whether translational repression and deadenylation are coupled or represent independent repressive mechanisms is unclear. Another remaining question is whether translational repression also requires GW182 proteins to interact with both PABP and deadenylases. To address these questions, we characterized the interaction of Drosophila melanogaster GW182 with deadenylases and defined the minimal requirements for a functional GW182 protein. Functional assays in D. melanogaster and human cells indicate that miRNA-mediated translational repression and degradation are mechanistically linked and are triggered through the interactions of GW182 proteins with PABP and deadenylases.


Subject(s)
Drosophila Proteins/metabolism , MicroRNAs/metabolism , Poly(A)-Binding Proteins/metabolism , RNA Interference , Ribonucleases/metabolism , Animals , Carrier Proteins/metabolism , Drosophila Proteins/chemistry , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , HeLa Cells , Humans , Protein Biosynthesis , Protein Interaction Domains and Motifs , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , Transcription Factors/chemistry
20.
Nature ; 492(7429): 376-81, 2012 Dec 20.
Article in English | MEDLINE | ID: mdl-23222520

ABSTRACT

In mammals, enlargement of the heart during embryonic development is primarily dependent on the increase in cardiomyocyte numbers. Shortly after birth, however, cardiomyocytes stop proliferating and further growth of the myocardium occurs through hypertrophic enlargement of the existing myocytes. As a consequence of the minimal renewal of cardiomyocytes during adult life, repair of cardiac damage through myocardial regeneration is very limited. Here we show that the exogenous administration of selected microRNAs (miRNAs) markedly stimulates cardiomyocyte proliferation and promotes cardiac repair. We performed a high-content microscopy, high-throughput functional screening for human miRNAs that promoted neonatal cardiomyocyte proliferation using a whole-genome miRNA library. Forty miRNAs strongly increased both DNA synthesis and cytokinesis in neonatal mouse and rat cardiomyocytes. Two of these miRNAs (hsa-miR-590 and hsa-miR-199a) were further selected for testing and were shown to promote cell cycle re-entry of adult cardiomyocytes ex vivo and to promote cardiomyocyte proliferation in both neonatal and adult animals. After myocardial infarction in mice, these miRNAs stimulated marked cardiac regeneration and almost complete recovery of cardiac functional parameters. The miRNAs identified hold great promise for the treatment of cardiac pathologies consequent to cardiomyocyte loss.


Subject(s)
MicroRNAs/analysis , MicroRNAs/genetics , Myocardium/cytology , Regeneration/genetics , Animals , Cell Proliferation , Cytokinesis , DNA/biosynthesis , Down-Regulation , Gene Library , Genetic Therapy , Heart/growth & development , Humans , Mice , MicroRNAs/therapeutic use , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Myocardial Infarction/therapy , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...