Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Methods Mol Biol ; 2784: 3-23, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38502475

RESUMO

In this chapter, we describe in detail how to perform a successful smFISH experiment and how to quantify mRNA transcripts in bacterial cells. The flexibility of the method allows for straightforward adaptation to different bacterial species and experimental conditions. Thanks to the feasibility of the approach, the method can easily be adapted by other laboratories. Finally, we believe that this method has a great potential to generate insights into the complicated life of bacteria.


Assuntos
Bactérias , RNA , Hibridização in Situ Fluorescente/métodos , RNA Mensageiro/genética , Bactérias/genética
2.
Curr Biol ; 33(22): 4880-4892.e14, 2023 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-37879333

RESUMO

Bacteria undergo cycles of growth and starvation to which they must adapt swiftly. One important strategy for adjusting growth rates relies on ribosomal levels. Although high ribosomal levels are required for fast growth, their dynamics during starvation remain unclear. Here, we analyzed ribosomal RNA (rRNA) content of individual Salmonella cells by using fluorescence in situ hybridization (rRNA-FISH) and measured a dramatic decrease in rRNA numbers only in a subpopulation during nutrient limitation, resulting in a bimodal distribution of cells with high and low rRNA content. During nutritional upshifts, the two subpopulations were associated with distinct phenotypes. Using a transposon screen coupled with rRNA-FISH, we identified two mutants, DksA and RNase I, acting on rRNA transcription shutdown and degradation, which abolished the formation of the subpopulation with low rRNA content. Our work identifies a bacterial mechanism for regulation of ribosomal bimodality that may be beneficial for population survival during starvation.


Assuntos
RNA Ribossômico , Ribossomos , RNA Ribossômico/genética , Hibridização in Situ Fluorescente , Ribossomos/metabolismo , Salmonella/genética , Salmonella/metabolismo , Estresse Fisiológico
3.
Proc Natl Acad Sci U S A ; 120(28): e2218812120, 2023 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-37399397

RESUMO

Encounters between host cells and intracellular bacterial pathogens lead to complex phenotypes that determine the outcome of infection. Single-cell RNA sequencing (scRNA-seq) is increasingly used to study the host factors underlying diverse cellular phenotypes but has limited capacity to analyze the role of bacterial factors. Here, we developed scPAIR-seq, a single-cell approach to analyze infection with a pooled library of multiplex-tagged, barcoded bacterial mutants. Infected host cells and barcodes of intracellular bacterial mutants are both captured by scRNA-seq to functionally analyze mutant-dependent changes in host transcriptomes. We applied scPAIR-seq to macrophages infected with a library of Salmonella Typhimurium secretion system effector mutants. We analyzed redundancy between effectors and mutant-specific unique fingerprints and mapped the global virulence network of each individual effector by its impact on host immune pathways. ScPAIR-seq is a powerful tool to untangle bacterial virulence strategies and their complex interplay with host defense strategies that drive infection outcome.


Assuntos
Macrófagos , Salmonella typhimurium , Virulência/genética , Macrófagos/metabolismo , Fatores de Virulência/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno/genética
4.
Curr Opin Immunol ; 84: 102369, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37473458

RESUMO

Macrophages are phagocytic cells distributed across tissues that sustain homeostasis by constantly probing their local environment. Upon perturbations, macrophages rewire their energy metabolism to execute their immune programs. Intensive research in the field of immunometabolism highlights cell-intrinsic immunometabolites such as succinate and itaconate as immunomodulatory signals. A role for cell-extrinsic stimuli now emerges with evidence for signals that shape macrophages' metabolism in a tissue-specific manner. In this review, we will cover macrophage immunometabolism in the gut, a complex metabolic and immunologically active tissue. During homeostasis, gut macrophages are constantly exposed to pro-inflammatory ligands from the microbiota, and in contrast, are balanced by microbiota-derived anti-inflammatory metabolites. Given their extensive metabolic changes during activation, spatial analyses of the tissue will allow the characterization of metabolic niches of macrophage in the gut. Identifying metabolic perturbations of macrophage subsets during chronic inflammation and infection can direct future tissue-specific metabolotherapies.


Assuntos
Metabolismo Energético , Macrófagos , Humanos , Ácido Succínico/metabolismo , Imunidade , Inflamação
5.
Cell ; 186(12): 2690-2704.e20, 2023 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-37295405

RESUMO

Biofilm formation is generally recognized as a bacterial defense mechanism against environmental threats, including antibiotics, bacteriophages, and leukocytes of the human immune system. Here, we show that for the human pathogen Vibrio cholerae, biofilm formation is not only a protective trait but also an aggressive trait to collectively predate different immune cells. We find that V. cholerae forms biofilms on the eukaryotic cell surface using an extracellular matrix comprising primarily mannose-sensitive hemagglutinin pili, toxin-coregulated pili, and the secreted colonization factor TcpF, which differs from the matrix composition of biofilms on other surfaces. These biofilms encase immune cells and establish a high local concentration of a secreted hemolysin to kill the immune cells before the biofilms disperse in a c-di-GMP-dependent manner. Together, these results uncover how bacteria employ biofilm formation as a multicellular strategy to invert the typical relationship between human immune cells as the hunters and bacteria as the hunted.


Assuntos
Vibrio cholerae , Animais , Humanos , Vibrio cholerae/metabolismo , Comportamento Predatório , Biofilmes , Fímbrias Bacterianas , Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica
6.
Infect Immun ; 91(4): e0043822, 2023 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-36939328

RESUMO

Bacterial pathogens can invade the tissue and establish a protected intracellular niche at the site of invasion that can spread locally (e.g., microcolonies) or to systemic sites (e.g., granulomas). Invasion of the tissue and establishment of intracellular infection are rare events that are difficult to study in the in vivo setting but have critical clinical consequences, such as long-term carriage, reinfections, and emergence of antibiotic resistance. Here, I discuss Salmonella interactions with its host macrophage during early stages of infection and their critical role in determining infection outcome. The dynamics of host-pathogen interactions entail highly heterogenous host immunity, bacterial virulence, and metabolic cross talk, requiring in vivo analysis at single-cell resolution. I discuss models and single-cell approaches that provide a global understanding of the establishment of a protected intracellular niche within the tissue and the host-pathogen landscape at infection bottlenecks during early stages of infection. Studying cellular host-pathogen interactions in vivo can improve our knowledge of the trajectory of infection between the initial inoculation with a dose of pathogens and the appearance of symptoms of disease.


Assuntos
Macrófagos , Salmonella , Macrófagos/microbiologia , Bactérias , Interações Hospedeiro-Patógeno
7.
Elife ; 122023 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-36691727

RESUMO

Unicellular algae, termed phytoplankton, greatly impact the marine environment by serving as the basis of marine food webs and by playing central roles in the biogeochemical cycling of elements. The interactions between phytoplankton and heterotrophic bacteria affect the fitness of both partners. It is becoming increasingly recognized that metabolic exchange determines the nature of such interactions, but the underlying molecular mechanisms remain underexplored. Here, we investigated the molecular and metabolic basis for the bacterial lifestyle switch, from coexistence to pathogenicity, in Sulfitobacter D7 during its interaction with Emiliania huxleyi, a cosmopolitan bloom-forming phytoplankter. To unravel the bacterial lifestyle switch, we analyzed bacterial transcriptomes in response to exudates derived from algae in exponential growth and stationary phase, which supported the Sulfitobacter D7 coexistence and pathogenicity lifestyles, respectively. In pathogenic mode, Sulfitobacter D7 upregulated flagellar motility and diverse transport systems, presumably to maximize assimilation of E. huxleyi-derived metabolites released by algal cells upon cell death. Algal dimethylsulfoniopropionate (DMSP) was a pivotal signaling molecule that mediated the transition between the lifestyles, supporting our previous findings. However, the coexisting and pathogenic lifestyles were evident only in the presence of additional algal metabolites. Specifically, we discovered that algae-produced benzoate promoted the growth of Sulfitobacter D7 and hindered the DMSP-induced lifestyle switch to pathogenicity, demonstrating that benzoate is important for maintaining the coexistence of algae and bacteria. We propose that bacteria can sense the physiological state of the algal host through changes in the metabolic composition, which will determine the bacterial lifestyle during interaction.


Assuntos
Haptófitas , Rhodobacteraceae , Fitoplâncton/metabolismo , Fitoplâncton/microbiologia
8.
Methods Mol Biol ; 2427: 133-147, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35619031

RESUMO

Complex interactions between diverse host immune cells can determine the outcome of pathogen infections. Advances in single-cell RNA sequencing (scRNA-seq) allow detection of the transcriptional patterns of different immune cells at steady state and after infection. To reveal the complex interactions of the human immune system in response to diverse intracellular pathogens, we developed a protocol for scRNA-seq of ex vivo infected human peripheral blood mononuclear cells (PBMCs). We demonstrate here infection with Salmonella enterica serovar Typhimurium, but this protocol can be used for any other pathogen of interest, and expand our knowledge of human host-pathogen biology.


Assuntos
Interações Hospedeiro-Patógeno , Leucócitos Mononucleares , Interações Hospedeiro-Patógeno/genética , Humanos , Salmonella typhimurium/genética , Análise de Sequência de RNA/métodos
9.
Nature ; 606(7914): 570-575, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35614218

RESUMO

The lineage and developmental trajectory of a cell are key determinants of cellular identity. In the vascular system, endothelial cells (ECs) of blood and lymphatic vessels differentiate and specialize to cater to the unique physiological demands of each organ1,2. Although lymphatic vessels were shown to derive from multiple cellular origins, lymphatic ECs (LECs) are not known to generate other cell types3,4. Here we use recurrent imaging and lineage-tracing of ECs in zebrafish anal fins, from early development to adulthood, to uncover a mechanism of specialized blood vessel formation through the transdifferentiation of LECs. Moreover, we demonstrate that deriving anal-fin vessels from lymphatic versus blood ECs results in functional differences in the adult organism, uncovering a link between cell ontogeny and functionality. We further use single-cell RNA-sequencing analysis to characterize the different cellular populations and transition states involved in the transdifferentiation process. Finally, we show that, similar to normal development, the vasculature is rederived from lymphatics during anal-fin regeneration, demonstrating that LECs in adult fish retain both potency and plasticity for generating blood ECs. Overall, our research highlights an innate mechanism of blood vessel formation through LEC transdifferentiation, and provides in vivo evidence for a link between cell ontogeny and functionality in ECs.


Assuntos
Vasos Sanguíneos , Transdiferenciação Celular , Vasos Linfáticos , Nadadeiras de Animais/citologia , Animais , Vasos Sanguíneos/citologia , Linhagem da Célula , Células Endoteliais/citologia , Vasos Linfáticos/citologia , Peixe-Zebra
10.
Nat Microbiol ; 7(4): 497-507, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35365784

RESUMO

Following detection of bacteria, macrophages switch their metabolism from oxidative respiration through the tricarboxylic acid cycle to high-rate aerobic glycolysis. This immunometabolic shift enables pro-inflammatory and antimicrobial responses and is facilitated by the accumulation of fatty acids, tricarboxylic acid-derived metabolites and catabolism of amino acids. Recent studies have shown that these immunometabolites are co-opted by pathogens as environmental cues for expression of virulence genes. We review mechanisms by which host immunometabolites regulate bacterial pathogenicity and discuss opportunities for the development of therapeutics targeting metabolic host-pathogen crosstalk.


Assuntos
Infecções Bacterianas , Infecções Bacterianas/metabolismo , Ciclo do Ácido Cítrico , Humanos , Macrófagos/microbiologia , Oxirredução , Virulência
11.
Immunity ; 55(3): 442-458.e8, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35182483

RESUMO

Consecutive exposures to different pathogens are highly prevalent and often alter the host immune response. However, it remains unknown how a secondary bacterial infection affects an ongoing adaptive immune response elicited against primary invading pathogens. We demonstrated that recruitment of Sca-1+ monocytes into lymphoid organs during Salmonella Typhimurium (STm) infection disrupted pre-existing germinal center (GC) reactions. GC responses induced by influenza, plasmodium, or commensals deteriorated following STm infection. GC disruption was independent of the direct bacterial interactions with B cells and instead was induced through recruitment of CCR2-dependent Sca-1+ monocytes into the lymphoid organs. GC collapse was associated with impaired cellular respiration and was dependent on TNFα and IFNγ, the latter of which was essential for Sca-1+ monocyte differentiation. Monocyte recruitment and GC disruption also occurred during LPS-supplemented vaccination and Listeria monocytogenes infection. Thus, systemic activation of the innate immune response upon severe bacterial infection is induced at the expense of antibody-mediated immunity.


Assuntos
Infecções Bacterianas , Listeriose , Linfócitos B , Centro Germinativo , Humanos , Monócitos
12.
Immunity ; 54(12): 2712-2723.e6, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34788598

RESUMO

Interactions between intracellular bacteria and mononuclear phagocytes give rise to diverse cellular phenotypes that may determine the outcome of infection. Recent advances in single-cell RNA sequencing (scRNA-seq) have identified multiple subsets within the mononuclear population, but implications to their function during infection are limited. Here, we surveyed the mononuclear niche of intracellular Salmonella Typhimurium (S.Tm) during early systemic infection in mice. We described eclipse-like growth kinetics in the spleen, with a first phase of bacterial control mediated by tissue-resident red-pulp macrophages. A second phase involved extensive bacterial replication within a macrophage population characterized by CD9 expression. We demonstrated that CD9+ macrophages induced pathways for detoxificating oxidized lipids, that may be utilized by intracellular S.Tm. We established that CD9+ macrophages originated from non-classical monocytes (NCM), and NCM-depleted mice were more resistant to S.Tm infection. Our study defines macrophage subset-specific host-pathogen interactions that determine early infection dynamics and infection outcome of the entire organism.


Assuntos
Macrófagos/imunologia , Infecções por Salmonella/imunologia , Salmonella typhimurium/fisiologia , Baço/imunologia , Animais , Interações Hospedeiro-Patógeno , Humanos , Espaço Intracelular , Metabolismo dos Lipídeos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oxirredução , Análise de Célula Única , Baço/microbiologia , Tetraspanina 29/metabolismo
13.
Science ; 371(6527): 400-405, 2021 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-33479153

RESUMO

Key to the success of intracellular pathogens is the ability to sense and respond to a changing host cell environment. Macrophages exposed to microbial products undergo metabolic changes that drive inflammatory responses. However, the role of macrophage metabolic reprogramming in bacterial adaptation to the intracellular environment has not been explored. Here, using metabolic profiling and dual RNA sequencing, we show that succinate accumulation in macrophages is sensed by intracellular Salmonella Typhimurium (S. Tm) to promote antimicrobial resistance and type III secretion. S Tm lacking the succinate uptake transporter DcuB displays impaired survival in macrophages and in mice. Thus, S Tm co-opts the metabolic reprogramming of infected macrophages as a signal that induces its own virulence and survival, providing an additional perspective on metabolic host-pathogen cross-talk.


Assuntos
Interações Hospedeiro-Patógeno , Macrófagos/metabolismo , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidade , Ácido Succínico/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sobrevivência Celular , Transportadores de Ácidos Dicarboxílicos/genética , Transportadores de Ácidos Dicarboxílicos/metabolismo , Modelos Animais de Doenças , Feminino , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , RNA-Seq , Salmonella typhimurium/genética , Virulência
14.
Sci Rep ; 9(1): 19244, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31848386

RESUMO

Dual transcriptional profiling of host and bacteria during infection is challenging due to the low abundance of bacterial mRNA. We report Pathogen Hybrid Capture (PatH-Cap), a method to enrich for bacterial mRNA and deplete bacterial rRNA simultaneously from dual RNA-seq libraries using transcriptome-specific probes. By addressing both the differential RNA content of the host relative to the infecting bacterium and the overwhelming abundance of uninformative structural RNAs (rRNA, tRNA) of both species in a single step, this approach enables analysis of very low-input RNA samples. By sequencing libraries before (pre-PatH-Cap) and after (post-PatH-Cap) enrichment, we achieve dual transcriptional profiling of host and bacteria, respectively, from the same sample. Importantly, enrichment preserves relative transcript abundance and increases the number of unique bacterial transcripts per gene in post-PatH-Cap libraries compared to pre-PatH-Cap libraries at the same sequencing depth, thereby decreasing the sequencing depth required to fully capture the transcriptional profile of the infecting bacteria. We demonstrate that PatH-Cap enables the study of low-input samples including single eukaryotic cells infected by 1-3 Pseudomonas aeruginosa bacteria and paired host-pathogen temporal gene expression analysis of Mycobacterium tuberculosis infecting macrophages. PatH-Cap can be applied to the study of a range of pathogens and microbial species, and more generally, to lowly-abundant species in mixed populations.


Assuntos
Perfilação da Expressão Gênica , Interações Hospedeiro-Parasita , Mycobacterium tuberculosis/fisiologia , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/fisiologia , RNA Bacteriano , RNA Mensageiro , Tuberculose/metabolismo , Animais , Camundongos , Hibridização de Ácido Nucleico , Infecções por Pseudomonas/patologia , RNA Bacteriano/química , RNA Bacteriano/isolamento & purificação , RNA Bacteriano/metabolismo , RNA Mensageiro/química , RNA Mensageiro/isolamento & purificação , RNA Mensageiro/metabolismo , Tuberculose/patologia
15.
Nat Commun ; 10(1): 3266, 2019 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-31332193

RESUMO

Complex interactions between different host immune cell types can determine the outcome of pathogen infections. Advances in single cell RNA-sequencing (scRNA-seq) allow probing of these immune interactions, such as cell-type compositions, which are then interpreted by deconvolution algorithms using bulk RNA-seq measurements. However, not all aspects of immune surveillance are represented by current algorithms. Here, using scRNA-seq of human peripheral blood cells infected with Salmonella, we develop a deconvolution algorithm for inferring cell-type specific infection responses from bulk measurements. We apply our dynamic deconvolution algorithm to a cohort of healthy individuals challenged ex vivo with Salmonella, and to three cohorts of tuberculosis patients during different stages of disease. We reveal cell-type specific immune responses associated not only with ex vivo infection phenotype but also with clinical disease stage. We propose that our approach provides a predictive power to identify risk for disease, and human infection outcomes.


Assuntos
Perfilação da Expressão Gênica/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Sistema Imunitário/metabolismo , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Algoritmos , Células Cultivadas , Análise por Conglomerados , Estudos de Coortes , Interações Hospedeiro-Patógeno/genética , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/microbiologia , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Células T Matadoras Naturais/microbiologia , Valor Preditivo dos Testes , Salmonella/fisiologia , Infecções por Salmonella/genética , Infecções por Salmonella/microbiologia
16.
Curr Opin Microbiol ; 42: 31-39, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29049916

RESUMO

Despite the availability of antibiotics and immunization, infectious diseases remain a major cause of malignancy and death worldwide. Yet, it is well documented that for most infectious agents, clinical disease develops in only a small minority of infected individuals. There is, in fact, great heterogeneity in infection outcome, from complete clearance of the pathogen to severe illness. Understanding this variation remains elusive, despite its great potential to equip us with new tools for the treatment of infectious diseases. Here, we propose a novel perspective for studying this diversity in human infection outcome, one that utilizes single-cell analysis technologies. Recent advances in single-cell RNA-seq technologies allow the detection of rare subpopulations that play important roles in host-pathogen interactions. We propose that applying single-cell RNA-seq to the study of infection can provide a 'fingerprint' of the immune cell types that are associated with the ability of the host to clear a pathogen and, thereby, broaden our current understanding of variation in susceptibility to infection within the population.


Assuntos
Doenças Transmissíveis/imunologia , Interações Hospedeiro-Patógeno/imunologia , Sistema Imunitário/citologia , Antibacterianos/efeitos adversos , Antibacterianos/uso terapêutico , Suscetibilidade a Doenças/imunologia , Suscetibilidade a Doenças/microbiologia , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Análise de Célula Única/métodos
17.
Genome Biol ; 18(1): 200, 2017 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-29073931

RESUMO

The interaction between a pathogen and a host is a highly dynamic process in which both agents activate complex programs. Here, we introduce a single-cell RNA-sequencing method, scDual-Seq, that simultaneously captures both host and pathogen transcriptomes. We use it to study the process of infection of individual mouse macrophages with the intracellular pathogen Salmonella typhimurium. Among the infected macrophages, we find three subpopulations and we show evidence for a linear progression through these subpopulations, supporting a model in which these three states correspond to consecutive stages of infection.


Assuntos
Perfilação da Expressão Gênica/métodos , Macrófagos/metabolismo , Macrófagos/microbiologia , Salmonella typhimurium/genética , Análise de Sequência de RNA/métodos , Animais , Regulação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Análise de Célula Única
18.
Science ; 357(6356): 1156-1160, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28912244

RESUMO

Growing evidence suggests that microbes can influence the efficacy of cancer therapies. By studying colon cancer models, we found that bacteria can metabolize the chemotherapeutic drug gemcitabine (2',2'-difluorodeoxycytidine) into its inactive form, 2',2'-difluorodeoxyuridine. Metabolism was dependent on the expression of a long isoform of the bacterial enzyme cytidine deaminase (CDDL), seen primarily in Gammaproteobacteria. In a colon cancer mouse model, gemcitabine resistance was induced by intratumor Gammaproteobacteria, dependent on bacterial CDDL expression, and abrogated by cotreatment with the antibiotic ciprofloxacin. Gemcitabine is commonly used to treat pancreatic ductal adenocarcinoma (PDAC), and we hypothesized that intratumor bacteria might contribute to drug resistance of these tumors. Consistent with this possibility, we found that of the 113 human PDACs that were tested, 86 (76%) were positive for bacteria, mainly Gammaproteobacteria.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/microbiologia , Desoxicitidina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/microbiologia , Animais , Neoplasias do Colo/microbiologia , Desoxicitidina/uso terapêutico , Gammaproteobacteria/isolamento & purificação , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mycoplasma hyorhinis/isolamento & purificação , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/microbiologia , Gencitabina , Neoplasias Pancreáticas
19.
PLoS Pathog ; 13(5): e1006363, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28505176

RESUMO

A key to the pathogenic success of Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, is the capacity to survive within host macrophages. Although several factors required for this survival have been identified, a comprehensive knowledge of such factors and how they work together to manipulate the host environment to benefit bacterial survival are not well understood. To systematically identify Mtb factors required for intracellular growth, we screened an arrayed, non-redundant Mtb transposon mutant library by high-content imaging to characterize the mutant-macrophage interaction. Based on a combination of imaging features, we identified mutants impaired for intracellular survival. We then characterized the phenotype of infection with each mutant by profiling the induced macrophage cytokine response. Taking a systems-level approach to understanding the biology of identified mutants, we performed a multiparametric analysis combining pathogen and host phenotypes to predict functional relationships between mutants based on clustering. Strikingly, mutants defective in two well-known virulence factors, the ESX-1 protein secretion system and the virulence lipid phthiocerol dimycocerosate (PDIM), clustered together. Building upon the shared phenotype of loss of the macrophage type I interferon (IFN) response to infection, we found that PDIM production and export are required for coordinated secretion of ESX-1-substrates, for phagosomal permeabilization, and for downstream induction of the type I IFN response. Multiparametric clustering also identified two novel genes that are required for PDIM production and induction of the type I IFN response. Thus, multiparametric analysis combining host and pathogen infection phenotypes can be used to identify novel functional relationships between genes that play a role in infection.


Assuntos
Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Mycobacterium tuberculosis/patogenicidade , Fagossomos/microbiologia , Tuberculose/microbiologia , Animais , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Linhagem Celular , Citocinas/imunologia , Citocinas/metabolismo , Biblioteca Gênica , Interações Hospedeiro-Patógeno , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Mutação , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/imunologia , Fagossomos/imunologia , Fenótipo , Tuberculose/imunologia , Virulência
20.
Curr Opin Microbiol ; 36: 69-75, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28214736

RESUMO

Most of our understanding of the host-bacterium interaction has come from studies of bulk populations. In reality, highly adaptable and dynamic host cells and bacteria engage in complex, diverse interactions. This complexity necessarily limits the depth of understanding that can be gained with bulk population measurements. Here, we will review the merit of single cell analysis to characterize this diversity that can trigger heterogeneous outcomes. We will discuss heterogeneity of bacterial and host populations, differences in host microenvironments, technological advances that facilitate the analysis of rare subpopulations, and the potential relevance of these subpopulations to infection outcomes. We focus our discussion on intracellular bacterial pathogens and on methods that characterize and quantify RNA in single cells, aiming to highlight how novel methodologies have the potential to characterize the multidimensional process of infection and to provide answers to some of the most fundamental questions in the field.


Assuntos
Interações Hospedeiro-Patógeno , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Bactérias/patogenicidade , Citoplasma/microbiologia , Humanos , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidade , RNA/análise , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA