Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Microsc ; 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38656474

RESUMO

Modern life science research is a collaborative effort. Few research groups can single-handedly support the necessary equipment, expertise and personnel needed for the ever-expanding portfolio of technologies that are required across multiple disciplines in today's life science endeavours. Thus, research institutes are increasingly setting up scientific core facilities to provide access and specialised support for cutting-edge technologies. Maintaining the momentum needed to carry out leading research while ensuring high-quality daily operations is an ongoing challenge, regardless of the resources allocated to establish such facilities. Here, we outline and discuss the range of activities required to keep things running once a scientific imaging core facility has been established. These include managing a wide range of equipment and users, handling repairs and service contracts, planning for equipment upgrades, renewals, or decommissioning, and continuously upskilling while balancing innovation and consolidation.

2.
Vet Res ; 55(1): 32, 2024 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-38493182

RESUMO

Outbreaks of West Nile virus (WNV) occur periodically, affecting both human and equine populations. There are no vaccines for humans, and those commercialised for horses do not have sufficient coverage. Specific antiviral treatments do not exist. Many drug discovery studies have been conducted, but since rodent or primate cell lines are normally used, results cannot always be transposed to horses. There is thus a need to develop relevant equine cellular models. Here, we used induced pluripotent stem cells to develop a new in vitro model of WNV-infected equine brain cells suitable for microplate assay, and assessed the cytotoxicity and antiviral activity of forty-one chemical compounds. We found that one nucleoside analog, 2'C-methylcytidine, blocked WNV infection in equine brain cells, whereas other compounds were either toxic or ineffective, despite some displaying anti-viral activity in human cell lines. We also revealed an unexpected proviral effect of statins in WNV-infected equine brain cells. Our results thus identify a potential lead for future drug development and underscore the importance of using a tissue- and species-relevant cellular model for assessing the activity of antiviral compounds.


Assuntos
Doenças dos Cavalos , Células-Tronco Pluripotentes Induzidas , Febre do Nilo Ocidental , Vírus do Nilo Ocidental , Animais , Cavalos , Humanos , Febre do Nilo Ocidental/veterinária , Febre do Nilo Ocidental/epidemiologia , Encéfalo , Antivirais/farmacologia , Antivirais/uso terapêutico , Doenças dos Cavalos/tratamento farmacológico
3.
J Infect Dis ; 225(6): 1005-1010, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-32582947

RESUMO

The bacterial pathogen Listeria monocytogenes invades host cells, ruptures the internalization vacuole, and reaches the cytosol for replication. A high-content small interfering RNA (siRNA) microscopy screen allowed us to identify epithelial cell factors involved in L. monocytogenes vacuolar rupture, including the serine/threonine kinase Taok2. Kinase activity inhibition using a specific drug validated a role for Taok2 in favoring L. monocytogenes cytoplasmic access. Furthermore, we showed that Taok2 recruitment to L. monocytogenes vacuoles requires the presence of pore-forming toxin listeriolysin O. Overall, our study identified the first set of host factors modulating L. monocytogenes vacuolar rupture and cytoplasmic access in epithelial cells.


Assuntos
Listeria monocytogenes , Listeriose , Proteínas de Bactérias , Citoplasma , Citosol , Proteínas Hemolisinas , Humanos , Listeriose/microbiologia , Vacúolos/microbiologia , Vacúolos/fisiologia
4.
BMC Biol ; 19(1): 136, 2021 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-34215263

RESUMO

BACKGROUND: Quantitative imaging of epithelial tissues requires bioimage analysis tools that are widely applicable and accurate. In the case of imaging 3D tissues, a common preprocessing step consists of projecting the acquired 3D volume on a 2D plane mapping the tissue surface. While segmenting the tissue cells is amenable on 2D projections, it is still very difficult and cumbersome in 3D. However, for many specimen and models used in developmental and cell biology, the complex content of the image volume surrounding the epithelium in a tissue often reduces the visibility of the biological object in the projection, compromising its subsequent analysis. In addition, the projection may distort the geometry of the tissue and can lead to strong artifacts in the morphology measurement. RESULTS: Here we introduce a user-friendly toolbox built to robustly project epithelia on their 2D surface from 3D volumes and to produce accurate morphology measurement corrected for the projection distortion, even for very curved tissues. Our toolbox is built upon two components. LocalZProjector is a configurable Fiji plugin that generates 2D projections and height-maps from potentially large 3D stacks (larger than 40 GB per time-point) by only incorporating signal of the planes with local highest variance/mean intensity, despite a possibly complex image content. DeProj is a MATLAB tool that generates correct morphology measurements by combining the height-map output (such as the one offered by LocalZProjector) and the results of a cell segmentation on the 2D projection, hence effectively deprojecting the 2D segmentation in 3D. In this paper, we demonstrate their effectiveness over a wide range of different biological samples. We then compare its performance and accuracy against similar existing tools. CONCLUSIONS: We find that LocalZProjector performs well even in situations where the volume to project also contains unwanted signal in other layers. We show that it can process large images without a pre-processing step. We study the impact of geometrical distortions on morphological measurements induced by the projection. We measured very large distortions which are then corrected by DeProj, providing accurate outputs.


Assuntos
Imageamento Tridimensional , Microscopia
6.
Cell Rep ; 30(6): 1870-1882.e4, 2020 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-32049017

RESUMO

Aberrant macrophage activation during intracellular infection generates immunopathologies that can cause severe human morbidity. A better understanding of immune subversion strategies and macrophage phenotypic and functional responses is necessary to design host-directed intervention strategies. Here, we uncover a fine-tuned transcriptional response that is induced in primary and lesional macrophages infected by the parasite Leishmania amazonensis and dampens NF-κB and NLRP3 inflammasome activation. Subversion is amastigote-specific and characterized by a decreased expression of activating and increased expression of de-activating components of these pro-inflammatory pathways, thus revealing a regulatory dichotomy that abrogates the anti-microbial response. Changes in transcript abundance correlate with histone H3K9/14 hypoacetylation and H3K4 hypo-trimethylation in infected primary and lesional macrophages at promoters of NF-κB-related, pro-inflammatory genes. Our results reveal a Leishmania immune subversion strategy targeting host cell epigenetic regulation to establish conditions beneficial for parasite survival and open avenues for host-directed, anti-microbial drug discovery.


Assuntos
Histonas/metabolismo , Inflamassomos/metabolismo , Macrófagos/metabolismo , NF-kappa B/metabolismo , Animais , Leishmania
7.
Front Immunol ; 11: 569331, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33505391

RESUMO

The LabEx Milieu Interieur (MI) project is a clinical study centered on the detailed characterization of the baseline and induced immune responses in blood samples from 1,000 healthy donors. Analyses of these samples has lay ground for seminal studies on the genetic and environmental determinants of immunologic variance in a healthy cohort population. In the current study we developed in vitro methods enabling standardized quantification of MI-cohort-derived primary fibroblasts responses. Our results show that in vitro human donor cohort fibroblast responses to stimulation by different MAMPs analogs allows to characterize individual donor immune-phenotype variability. The results provide proof-of-concept foundation to a new experimental framework for such studies. A bio-bank of primary fibroblast lines was generated from 323 out of 1,000 healthy individuals selected from the MI-study cohort. To study inter-donor variability of innate immune response in primary human dermal fibroblasts we chose to measure the TLR3 and TLR4 response pathways, both receptors being expressed and previously studied in fibroblasts. We established high-throughput automation compatible methods for standardized primary fibroblast cell activation, using purified MAMPS analogs, poly I:C and LPS that stimulate TLR3 and TLR4 pathways respectively. These results were in turn compared with a stimulation method using infection by HSV-1 virus. Our "Add-only" protocol minimizes high-throughput automation system variability facilitating whole process automation from cell plating through stimulation to recovery of cell supernatants, and fluorescent labeling. Images were acquired automatically by high-throughput acquisition on an automated high-content imaging microscope. Under these methodological conditions standardized image acquisition provided for quantification of cellular responses allowing biological variability to be measured with low system noise and high biological signal fidelity. Optimal for automated analysis of immuno-phenotype of primary human cell responses our method and experimental framework as reported here is highly compatible to high-throughput screening protocols like those necessary for chemo-genomic screening. In context of primary fibroblasts derived from donors enrolled to the MI-clinical-study our results open the way to assert the utility of studying immune-phenotype characteristics relevant to a human clinical cohort.


Assuntos
Variação Biológica da População/imunologia , Fibroblastos/imunologia , Fibroblastos/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Bioensaio/métodos , Linhagem Celular , Células Cultivadas , Citocinas/metabolismo , Feminino , Citometria de Fluxo , Expressão Gênica , Genes Reporter , Herpesvirus Humano 1/imunologia , Humanos , Lipopolissacarídeos/imunologia , Pessoa de Meia-Idade , Poli I-C/imunologia , Polilisina/imunologia , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
8.
Trends Parasitol ; 35(7): 559-570, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31176583

RESUMO

Cell-based phenotypic screening has proven to be valuable, notably in recapitulating relevant biological conditions, for example, the host cell/pathogen niche. However, the corresponding methodological complexity is not readily compatible with high-throughput pipelines, and fails to inform either molecular target or mechanism of action, which frustrates conventional drug-discovery roadmaps. We review the state-of-the-art and emerging technologies that suggest new strategies for harnessing value from the complexity of phenotypic screening and augmenting powerful utility for translational drug discovery. Advances in cellular, molecular, and bioinformatics technologies are converging at a cutting edge where the complexity of phenotypic screening may no longer be considered a hinderance but rather a catalyst to chemotherapeutic discovery for infectious diseases.


Assuntos
Doenças Transmissíveis/tratamento farmacológico , Biologia Computacional/tendências , Descoberta de Drogas/métodos , Células Cultivadas , Interações Hospedeiro-Patógeno , Humanos , Fenótipo
9.
Curr Opin Microbiol ; 43: 193-198, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29567588

RESUMO

We consider in review current state-of-the-art fluorescence microscopy for investigating the host-pathogen interface. Our perspective is honed from years with literally thousands of microbiologists using the variety of imaging technologies available within our dedicated BSL2/BSL3 optical imaging research service facilities at the Institut Pasteur Paris founded from scratch in 2001. During fifteen years learning from the success and failures of introducing different fluorescence imaging technologies, methods, and technical development strategies we provide here a synopsis review of our experience to date and a synthesis of how we see the future in perspective for fluorescence imaging at the host-pathogen interface.


Assuntos
Interações Hospedeiro-Patógeno , Microscopia de Fluorescência/métodos , Automação Laboratorial , Contenção de Riscos Biológicos , Humanos , Laboratórios/organização & administração , Microscopia de Fluorescência/instrumentação , Imagem Molecular/instrumentação , Imagem Molecular/métodos
10.
Methods Mol Biol ; 1535: 173-195, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27914079

RESUMO

Listeria monocytogenes is a bacterial pathogen which invades and multiplies within non-professional phagocytes. Signaling cascades involved in cellular entry have been extensively analyzed, but the events leading to vacuolar escape remain less clear. In this chapter, we detail a microscopy FRET-based assay which allows quantitatively measuring L. monocytogenes infection and escape from its internalization vacuole, as well as a correlative light/electron microscopy method to investigate the morphological features of the vacuolar compartments containing L. monocytogenes.


Assuntos
Transferência Ressonante de Energia de Fluorescência/métodos , Listeria monocytogenes/metabolismo , Listeria monocytogenes/ultraestrutura , Microscopia Eletrônica/métodos , Microscopia de Fluorescência/métodos , Vacúolos/metabolismo , Transporte Biológico , Vacúolos/ultraestrutura
11.
Nat Rev Drug Discov ; 15(11): 751-769, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27616293

RESUMO

The common and persistent failures to translate promising preclinical drug candidates into clinical success highlight the limited effectiveness of disease models currently used in drug discovery. An apparent reluctance to explore and adopt alternative cell- and tissue-based model systems, coupled with a detachment from clinical practice during assay validation, contributes to ineffective translational research. To help address these issues and stimulate debate, here we propose a set of principles to facilitate the definition and development of disease-relevant assays, and we discuss new opportunities for exploiting the latest advances in cell-based assay technologies in drug discovery, including induced pluripotent stem cells, three-dimensional (3D) co-culture and organ-on-a-chip systems, complemented by advances in single-cell imaging and gene editing technologies. Funding to support precompetitive, multidisciplinary collaborations to develop novel preclinical models and cell-based screening technologies could have a key role in improving their clinical relevance, and ultimately increase clinical success rates.


Assuntos
Técnicas de Cultura de Células/métodos , Descoberta de Drogas/métodos , Modelos Biológicos , Animais , Linhagem Celular Transformada , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala/métodos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/fisiologia , Preparações Farmacêuticas/administração & dosagem
12.
Antimicrob Agents Chemother ; 60(5): 2822-33, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26902771

RESUMO

Existing therapies for leishmaniases present significant limitations, such as toxic side effects, and are rendered inefficient by parasite resistance. It is of utmost importance to develop novel drugs targeting Leishmania that take these two limitations into consideration. We thus chose a target-based approach using an exoprotein kinase, Leishmania casein kinase 1.2 (LmCK1.2) that was recently shown to be essential for intracellular parasite survival and infectivity. We developed a four-step pipeline to identify novel selective antileishmanial compounds. In step 1, we screened 5,018 compounds from kinase-biased libraries with Leishmania and mammalian CK1 in order to identify hit compounds and assess their specificity. For step 2, we selected 88 compounds among those with the lowest 50% inhibitory concentration to test their biological activity on host-free parasites using a resazurin reduction assay and on intramacrophagic amastigotes using a high content phenotypic assay. Only 75 compounds showed antileishmanial activity and were retained for step 3 to evaluate their toxicity against mouse macrophages and human cell lines. The four compounds that displayed a selectivity index above 10 were then assessed for their affinity to LmCK1.2 using a target deconvolution strategy in step 4. Finally, we retained two compounds, PP2 and compound 42, for which LmCK1.2 seems to be the primary target. Using this four-step pipeline, we identify from several thousand molecules, two lead compounds with a selective antileishmanial activity.


Assuntos
Antiprotozoários/farmacologia , Leishmania/efeitos dos fármacos , Animais , Antiprotozoários/química , Caseína Quinase I/metabolismo , Linhagem Celular , Descoberta de Drogas , Humanos , Leishmania/metabolismo , Macrófagos/parasitologia , Isoformas de Proteínas/metabolismo
13.
Appl Environ Microbiol ; 82(1): 211-7, 2016 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-26497455

RESUMO

Listeria monocytogenes is a Gram-positive bacterium and a facultative intracellular pathogen that invades mammalian cells, disrupts its internalization vacuole, and proliferates in the host cell cytoplasm. Here, we describe a novel image-based microscopy assay that allows discrimination between cellular entry and vacuolar escape, enabling high-content screening to identify factors specifically involved in these two steps. We first generated L. monocytogenes and Listeria innocua strains expressing a ß-lactamase covalently attached to the bacterial cell wall. These strains were then incubated with HeLa cells containing the Förster resonance energy transfer (FRET) probe CCF4 in their cytoplasm. The CCF4 probe was cleaved by the bacterial surface ß-lactamase only in cells inoculated with L. monocytogenes but not those inoculated with L. innocua, thereby demonstrating bacterial access to the host cytoplasm. Subsequently, we performed differential immunofluorescence staining to distinguish extracellular versus total bacterial populations in samples that were also analyzed by the FRET-based assay. With this two-step analysis, bacterial entry can be distinguished from vacuolar rupture in a single experiment. Our novel approach represents a powerful tool for identifying factors that determine the intracellular niche of L. monocytogenes.


Assuntos
Citoplasma/microbiologia , Interações Hospedeiro-Patógeno , Listeria monocytogenes/fisiologia , Vacúolos/microbiologia , Proteínas de Bactérias/metabolismo , Transferência Ressonante de Energia de Fluorescência , Células HeLa , Compostos Heterocíclicos de 4 ou mais Anéis/metabolismo , Humanos , Listeria/enzimologia , Listeria/metabolismo , Listeria monocytogenes/enzimologia , Microscopia de Fluorescência , beta-Lactamases/metabolismo
14.
Cell Host Microbe ; 16(4): 517-30, 2014 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-25299335

RESUMO

Shigella enters epithlial cells via internalization into a vacuole. Subsequent vacuolar membrane rupture allows bacterial escape into the cytosol for replication and cell-to-cell spread. Bacterial effectors such as IpgD, a PI(4,5)P2 phosphatase that generates PI(5)P and alters host actin, facilitate this internalization. Here, we identify host proteins involved in Shigella uptake and vacuolar membrane rupture by high-content siRNA screening and subsequently focus on Rab11, a constituent of the recycling compartment. Rab11-positive vesicles are recruited to the invasion site before vacuolar rupture, and Rab11 knockdown dramatically decreases vacuolar membrane rupture. Additionally, Rab11 recruitment is absent and vacuolar rupture is delayed in the ipgD mutant that does not dephosphorylate PI(4,5)P2 into PI(5)P. Ultrastructural analyses of Rab11-positive vesicles further reveal that ipgD mutant-containing vacuoles become confined in actin structures that likely contribute to delayed vacular rupture. These findings provide insight into the underlying molecular mechanism of vacuole progression and rupture during Shigella invasion.


Assuntos
Proteínas de Bactérias/metabolismo , Citoplasma/microbiologia , Endocitose , Células Epiteliais/microbiologia , Monoéster Fosfórico Hidrolases/metabolismo , Shigella/fisiologia , Vacúolos/microbiologia , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas de Bactérias/genética , Células Epiteliais/fisiologia , Técnicas de Inativação de Genes , Interações Hospedeiro-Patógeno , Membranas Intracelulares/metabolismo , Monoéster Fosfórico Hidrolases/genética , Shigella/genética , Shigella/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
15.
FASEB J ; 28(2): 603-14, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24145721

RESUMO

The Notch signaling pathway is involved in liver development and regeneration. Here, we investigate the role of the 4 mammalian Notch paralogs in the regulation of hepatoblast proliferation and hepatocytic differentiation. Our model is based on bipotential mouse embryonic liver (BMEL) progenitors that can differentiate into hepatocytes or cholangiocytes in vitro and in vivo. BMEL cells were subjected to Notch antagonists or agonists. Blocking Notch activation with a γ-secretase inhibitor, at 50 µM for 48 h, reduced cell growth by 50%. S-phase entry was impaired, but no apoptosis was induced. A systematic paralog-specific strategy was set using lentiviral transduction with constitutively active forms of each Notch receptor along with inhibition of endogenous Notch signaling. This assay demonstrates that proliferation of BMEL cells requires Notch2 and Notch4 activity, resulting in significant down-regulation of p27(Kip1) and p57(Kip2) cyclin-dependent kinase inhibitors. Conversely, Notch3-expressing cells proliferate less and express 3-fold higher levels of p57(Kip2). The Notch3 cells present a hepatocyte-like morphology, enhanced multinucleation, and a ploidy shift. Moreover, Notch3 activity is conducive to hepatocytic differentiation in vitro, while its paralogs impede this fate. Our study provides the first evidence of a functional diversity among the mammalian Notch homologues in the proliferation and hepatocytic-lineage commitment of liver progenitors.


Assuntos
Hepatócitos/citologia , Hepatócitos/metabolismo , Fígado/citologia , Fígado/metabolismo , Receptores Notch/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Proliferação de Células , Imunofluorescência , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Receptores Notch/genética
16.
Antimicrob Agents Chemother ; 58(3): 1501-15, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24366737

RESUMO

Protein kinase inhibitors have emerged as new drugs in various therapeutic areas, including leishmaniasis, an important parasitic disease. Members of the Leishmania casein kinase 1 (CK1) family represent promising therapeutic targets. Leishmania casein kinase 1 isoform 2 (CK1.2) has been identified as an exokinase capable of phosphorylating host proteins, thus exerting a potential immune-suppressive action on infected host cells. Moreover, its inhibition reduces promastigote growth. Despite these important properties, its requirement for intracellular infection and its chemical validation as a therapeutic target in the disease-relevant amastigote stage remain to be established. In this study, we used a multidisciplinary approach combining bioinformatics, biochemical, and pharmacological analyses with a macrophage infection assay to characterize and define Leishmania CK1.2 as a valid drug target. We show that recombinant and transgenic Leishmania CK1.2 (i) can phosphorylate CK1-specific substrates, (ii) is sensitive to temperature, and (iii) is susceptible to CK1-specific inhibitors. CK1.2 is constitutively expressed at both the promastigote insect stage and the vertebrate amastigote stage. We further demonstrated that reduction of CK1 activity by specific inhibitors, such as D4476, blocks promastigote growth, strongly compromises axenic amastigote viability, and decreases the number of intracellular Leishmania donovani and L. amazonensis amastigotes in infected macrophages. These results underline the potential role of CK1 kinases in intracellular survival. The identification of differences in structure and inhibition profiles compared to those of mammalian CK1 kinases opens new opportunities for Leishmania CK1.2 antileishmanial drug development. Our report provides the first chemical validation of Leishmania CK1 protein kinases, required for amastigote intracellular survival, as therapeutic targets.


Assuntos
Caseína Quinase I/efeitos dos fármacos , Leishmania donovani/efeitos dos fármacos , Animais , Benzamidas/farmacologia , Caseína Quinase I/antagonistas & inibidores , Caseína Quinase I/genética , Caseína Quinase I/fisiologia , Sequência Conservada/genética , Cricetinae/parasitologia , Feminino , Imidazóis/farmacologia , Indóis/farmacologia , Isoquinolinas/farmacologia , Leishmania donovani/enzimologia , Leishmania donovani/genética , Leishmania donovani/patogenicidade , Leishmania donovani/fisiologia , Leishmaniose Visceral/tratamento farmacológico , Leishmaniose Visceral/parasitologia , Macrófagos/parasitologia , Camundongos Endogâmicos C57BL , Floroglucinol/análogos & derivados , Floroglucinol/farmacologia , Alinhamento de Sequência , Tripanossomicidas/farmacologia
17.
PLoS Negl Trop Dis ; 7(4): e2154, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23593521

RESUMO

BACKGROUND/OBJECTIVES: Human leishmaniases are parasitic diseases causing severe morbidity and mortality. No vaccine is available and numerous factors limit the use of current therapies. There is thus an urgent need for innovative initiatives to identify new chemotypes displaying selective activity against intracellular Leishmania amastigotes that develop and proliferate inside macrophages, thereby causing the pathology of leishmaniasis. METHODOLOGY/PRINCIPAL FINDINGS: We have developed a biologically sound High Content Analysis assay, based on the use of homogeneous populations of primary mouse macrophages hosting Leishmania amazonensis amastigotes. In contrast to classical promastigote-based screens, our assay more closely mimics the environment where intracellular amastigotes are growing within acidic parasitophorous vacuoles of their host cells. This multi-parametric assay provides quantitative data that accurately monitors the parasitic load of amastigotes-hosting macrophage cultures for the discovery of leishmanicidal compounds, but also their potential toxic effect on host macrophages. We validated our approach by using a small set of compounds of leishmanicidal drugs and recently published chemical entities. Based on their intramacrophagic leishmanicidal activity and their toxicity against host cells, compounds were classified as irrelevant or relevant for entering the next step in the drug discovery pipeline. CONCLUSIONS/SIGNIFICANCE: Our assay represents a new screening platform that overcomes several limitations in anti-leishmanial drug discovery. First, the ability to detect toxicity on primary macrophages allows for discovery of compounds able to cross the membranes of macrophage, vacuole and amastigote, thereby accelerating the hit to lead development process for compounds selectively targeting intracellular parasites. Second, our assay allows discovery of anti-leishmanials that interfere with biological functions of the macrophage required for parasite development and growth, such as organelle trafficking/acidification or production of microbicidal effectors. These data thus validate a novel phenotypic screening assay using virulent Leishmania amastigotes growing inside primary macrophage to identify new chemical entities with bona fide drug potential.


Assuntos
Antiprotozoários/farmacologia , Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Leishmania/patogenicidade , Macrófagos/parasitologia , Animais , Células Cultivadas , Leishmania/efeitos dos fármacos , Leishmaniose/parasitologia , Camundongos
18.
J Virol ; 84(5): 2282-93, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20015995

RESUMO

The HIV-1 Nef protein is a pathogenic factor modulating the behavior of infected cells. Nef induces actin cytoskeleton changes and impairs cell migration toward chemokines. We further characterized the morphology, cytoskeleton dynamics, and motility of HIV-1-infected lymphocytes. By using scanning electron microscopy, confocal immunofluorescence microscopy, and ImageStream technology, which combines flow cytometry and automated imaging, we report that HIV-1 induces a characteristic remodeling of the actin cytoskeleton. In infected lymphocytes, ruffle formation is inhibited, whereas long, thin filopodium-like protrusions are induced. Cells infected with HIV with nef deleted display a normal phenotype, and Nef expression alone, in the absence of other viral proteins, induces morphological changes. We also used an innovative imaging system to immobilize and visualize living individual cells in suspension. When combined with confocal "axial tomography," this technique greatly enhances three-dimensional optical resolution. With this technique, we confirmed the induction of long filopodium-like structures in unfixed Nef-expressing lymphocytes. The cytoskeleton reorganization induced by Nef is associated with an important impairment of cell movements. The adhesion and spreading of infected cells to fibronectin, their spontaneous motility, and their migration toward chemokines (CXCL12, CCL3, and CCL19) were all significantly decreased. Therefore, Nef induces complex effects on the lymphocyte actin cytoskeleton and cellular morphology, which likely impacts the capacity of infected cells to circulate and to encounter and communicate with bystander cells.


Assuntos
Movimento Celular/fisiologia , Extensões da Superfície Celular/metabolismo , Linfócitos/citologia , Linfócitos/virologia , Pseudópodes/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Forma Celular , Extensões da Superfície Celular/ultraestrutura , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Citometria de Fluxo/instrumentação , Citometria de Fluxo/métodos , Infecções por HIV/metabolismo , HIV-1/metabolismo , Humanos , Imageamento Tridimensional/instrumentação , Imageamento Tridimensional/métodos , Linfócitos/metabolismo , Pseudópodes/ultraestrutura , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
19.
PLoS One ; 4(10): e7375, 2009 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-19823575

RESUMO

Pathogens use diverse molecular machines to penetrate host cells and manipulate intracellular vesicular trafficking. Viruses employ glycoproteins, functionally and structurally similar to the SNARE proteins, to induce eukaryotic membrane fusion. Intracellular pathogens, on the other hand, need to block fusion of their infectious phagosomes with various endocytic compartments to escape from the degradative pathway. The molecular details concerning the mechanisms underlying this process are lacking. Using both an in vitro liposome fusion assay and a cellular assay, we showed that SNARE-like bacterial proteins block membrane fusion in eukaryotic cells by directly inhibiting SNARE-mediated membrane fusion. More specifically, we showed that IncA and IcmG/DotF, two SNARE-like proteins respectively expressed by Chlamydia and Legionella, inhibit the endocytic SNARE machinery. Furthermore, we identified that the SNARE-like motif present in these bacterial proteins encodes the inhibitory function. This finding suggests that SNARE-like motifs are capable of specifically manipulating membrane fusion in a wide variety of biological environments. Ultimately, this motif may have been selected during evolution because it is an efficient structural motif for modifying eukaryotic membrane fusion and thus contribute to pathogen survival.


Assuntos
Bactérias/metabolismo , Proteínas SNARE/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Transporte Biológico , Linhagem Celular , Chlamydia/metabolismo , Endocitose , Glicoproteínas/metabolismo , Legionella/metabolismo , Lipossomos/metabolismo , Microscopia Confocal/métodos , Modelos Biológicos , Fosfoproteínas/metabolismo , Ratos
20.
Methods Mol Biol ; 486: 29-41, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19347614

RESUMO

To understand signaling pathways in mammalian cells, cell-based assays are relatively new and extremely powerful tools. We have developed a battery of phenotypic assays to study signaling; two of them are described in detail in this chapter. A subset of these assays monitors mitogen-activated protein (MAP) kinase pathways. MAP kinases are principal regulators of fundamental processes in mammalian cells, including growth, cell division, differentiation, stress responses, and neoplastic transformation. Here we describe two cell-based assays querying the function of ERK (extracellular signal regulated kinase), one of the three principal MAP kinases in mammalian cells. We selected human umbilical vein endothelial cells (HUVECs), a primary cell type, because they show a very dynamic response to various activators. Both assays are phenotypic assays and use well-established phosphorylation-specific primary antibodies to study activation. Fluorochrome-coupled secondary antibodies were used to label phosphorylated target proteins; images were captured with the INCell Analyzer 3000 and analyzed with the INCell Analyzer 3000 software. The first of these two assays monitors phosphorylation of ERK1/2, while the second assay monitors activation of the transcription factor CREB (cAMP response element-binding protein). The assays described in this chapter cover major checkpoints of the ERK signaling pathway: (1) MAP kinase activation and (2) subsequent transcription factor activation. Both assays exhibit robust performance and can easily be used for high-throughput screening.


Assuntos
Bioensaio/métodos , Endotélio Vascular/enzimologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Transdução de Sinais , Células Cultivadas , Endotélio Vascular/efeitos dos fármacos , Humanos , Fosforilação/efeitos dos fármacos , Proteína Quinase C/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Veias Umbilicais/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...