Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 44
Filtrer
1.
STAR Protoc ; 5(3): 103137, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38878285

RÉSUMÉ

Ribosome quantification in single cells is typically achieved through fluorescence tagging of ribosomal proteins. Here, we present a protocol for comparing ribosomal levels in bacteria at different growth stages using fluorescence in situ hybridization of rRNA (rRNA-FISH), eliminating the need for genetic engineering of the strain of interest. We detail the steps for preparing bacterial samples, staining with fluorescent probes, and acquiring data using flow cytometry and microscopy. Furthermore, we provide guidelines on controlling for proper labeling through signal localization analysis. For complete details on the use and execution of this protocol, please refer to Ciolli Mattioli et al.1.

3.
Methods Mol Biol ; 2784: 3-23, 2024.
Article de Anglais | MEDLINE | ID: mdl-38502475

RÉSUMÉ

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.


Sujet(s)
Bactéries , ARN , Hybridation fluorescente in situ/méthodes , ARN messager/génétique , Bactéries/génétique
4.
Curr Biol ; 33(22): 4880-4892.e14, 2023 11 20.
Article de Anglais | MEDLINE | ID: mdl-37879333

RÉSUMÉ

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.


Sujet(s)
ARN ribosomique , Ribosomes , ARN ribosomique/génétique , Hybridation fluorescente in situ , Ribosomes/métabolisme , Salmonella/génétique , Salmonella/métabolisme , Stress physiologique
5.
Curr Opin Immunol ; 84: 102369, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37473458

RÉSUMÉ

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.


Sujet(s)
Métabolisme énergétique , Macrophages , Humains , Acide succinique/métabolisme , Immunité , Inflammation
6.
Proc Natl Acad Sci U S A ; 120(28): e2218812120, 2023 07 11.
Article de Anglais | MEDLINE | ID: mdl-37399397

RÉSUMÉ

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.


Sujet(s)
Macrophages , Salmonella typhimurium , Virulence/génétique , Macrophages/métabolisme , Facteurs de virulence/génétique , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Interactions hôte-pathogène/génétique
7.
Cell ; 186(12): 2690-2704.e20, 2023 06 08.
Article de Anglais | MEDLINE | ID: mdl-37295405

RÉSUMÉ

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.


Sujet(s)
Vibrio cholerae , Animaux , Humains , Vibrio cholerae/métabolisme , Comportement prédateur , Biofilms , Fimbriae bactériens , Protéines bactériennes/métabolisme , Régulation de l'expression des gènes bactériens
8.
Infect Immun ; 91(4): e0043822, 2023 04 18.
Article de Anglais | MEDLINE | ID: mdl-36939328

RÉSUMÉ

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.


Sujet(s)
Macrophages , Salmonella , Macrophages/microbiologie , Bactéries , Interactions hôte-pathogène
9.
Elife ; 122023 Jan 24.
Article de Anglais | MEDLINE | ID: mdl-36691727

RÉSUMÉ

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.


Sujet(s)
Haptophyta , Rhodobacteraceae , Phytoplancton/métabolisme , Phytoplancton/microbiologie
10.
Methods Mol Biol ; 2427: 133-147, 2022.
Article de Anglais | MEDLINE | ID: mdl-35619031

RÉSUMÉ

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.


Sujet(s)
Interactions hôte-pathogène , Agranulocytes , Interactions hôte-pathogène/génétique , Humains , Salmonella typhimurium/génétique , Analyse de séquence d'ARN/méthodes
11.
Nature ; 606(7914): 570-575, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35614218

RÉSUMÉ

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.


Sujet(s)
Vaisseaux sanguins , Transdifférenciation cellulaire , Vaisseaux lymphatiques , Nageoires animales/cytologie , Animaux , Vaisseaux sanguins/cytologie , Lignage cellulaire , Cellules endothéliales/cytologie , Vaisseaux lymphatiques/cytologie , Danio zébré
12.
Nat Microbiol ; 7(4): 497-507, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-35365784

RÉSUMÉ

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.


Sujet(s)
Infections bactériennes , Infections bactériennes/métabolisme , Cycle citrique , Humains , Macrophages/microbiologie , Oxydoréduction , Virulence
13.
Immunity ; 55(3): 442-458.e8, 2022 03 08.
Article de Anglais | MEDLINE | ID: mdl-35182483

RÉSUMÉ

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.


Sujet(s)
Infections bactériennes , Infections à Listeria , Lymphocytes B , Centre germinatif , Humains , Monocytes
14.
Immunity ; 54(12): 2712-2723.e6, 2021 12 14.
Article de Anglais | MEDLINE | ID: mdl-34788598

RÉSUMÉ

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.


Sujet(s)
Macrophages/immunologie , Salmonelloses/immunologie , Salmonella typhimurium/physiologie , Rate/immunologie , Animaux , Interactions hôte-pathogène , Humains , Espace intracellulaire , Métabolisme lipidique , Macrophages/microbiologie , Souris , Souris de lignée C57BL , Souris transgéniques , Oxydoréduction , Analyse sur cellule unique , Rate/microbiologie , Antigène CD9/métabolisme
15.
Science ; 371(6527): 400-405, 2021 01 22.
Article de Anglais | MEDLINE | ID: mdl-33479153

RÉSUMÉ

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.


Sujet(s)
Interactions hôte-pathogène , Macrophages/métabolisme , Salmonella typhimurium/métabolisme , Salmonella typhimurium/pathogénicité , Acide succinique/métabolisme , Systèmes de sécrétion de type III/métabolisme , Animaux , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Survie cellulaire , Transporteurs d'acides dicarboxyliques/génétique , Transporteurs d'acides dicarboxyliques/métabolisme , Modèles animaux de maladie humaine , Femelle , Macrophages/microbiologie , Souris , Souris de lignée C57BL , RNA-Seq , Salmonella typhimurium/génétique , Virulence
16.
Sci Rep ; 9(1): 19244, 2019 12 17.
Article de Anglais | MEDLINE | ID: mdl-31848386

RÉSUMÉ

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.


Sujet(s)
Analyse de profil d'expression de gènes , Interactions hôte-parasite , Mycobacterium tuberculosis/physiologie , Infections à Pseudomonas/métabolisme , Pseudomonas aeruginosa/physiologie , ARN bactérien , ARN messager , Tuberculose/métabolisme , Animaux , Souris , Hybridation d'acides nucléiques , Infections à Pseudomonas/anatomopathologie , ARN bactérien/composition chimique , ARN bactérien/isolement et purification , ARN bactérien/métabolisme , ARN messager/composition chimique , ARN messager/isolement et purification , ARN messager/métabolisme , Tuberculose/anatomopathologie
17.
Nat Commun ; 10(1): 3266, 2019 07 22.
Article de Anglais | MEDLINE | ID: mdl-31332193

RÉSUMÉ

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.


Sujet(s)
Analyse de profil d'expression de gènes/méthodes , Séquençage nucléotidique à haut débit/méthodes , Système immunitaire/métabolisme , Analyse de séquence d'ARN/méthodes , Analyse sur cellule unique/méthodes , Algorithmes , Cellules cultivées , Analyse de regroupements , Études de cohortes , Interactions hôte-pathogène/génétique , Humains , Système immunitaire/cytologie , Système immunitaire/microbiologie , Cellules T tueuses naturelles/immunologie , Cellules T tueuses naturelles/métabolisme , Cellules T tueuses naturelles/microbiologie , Valeur prédictive des tests , Salmonella/physiologie , Salmonelloses/génétique , Salmonelloses/microbiologie
18.
Curr Opin Microbiol ; 42: 31-39, 2018 04.
Article de Anglais | MEDLINE | ID: mdl-29049916

RÉSUMÉ

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.


Sujet(s)
Maladies transmissibles/immunologie , Interactions hôte-pathogène/immunologie , Système immunitaire/cytologie , Antibactériens/effets indésirables , Antibactériens/usage thérapeutique , Prédisposition aux maladies/immunologie , Prédisposition aux maladies/microbiologie , Séquençage nucléotidique à haut débit/méthodes , Humains , Analyse sur cellule unique/méthodes
19.
Genome Biol ; 18(1): 200, 2017 10 27.
Article de Anglais | MEDLINE | ID: mdl-29073931

RÉSUMÉ

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.


Sujet(s)
Analyse de profil d'expression de gènes/méthodes , Macrophages/métabolisme , Macrophages/microbiologie , Salmonella typhimurium/génétique , Analyse de séquence d'ARN/méthodes , Animaux , Régulation de l'expression des gènes , Souris , Souris de lignée C57BL , Analyse sur cellule unique
20.
Science ; 357(6356): 1156-1160, 2017 09 15.
Article de Anglais | MEDLINE | ID: mdl-28912244

RÉSUMÉ

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.


Sujet(s)
Antimétabolites antinéoplasiques/usage thérapeutique , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/microbiologie , Désoxycytidine/analogues et dérivés , Résistance aux médicaments antinéoplasiques , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/microbiologie , Animaux , Tumeurs du côlon/microbiologie , Désoxycytidine/usage thérapeutique , Gammaproteobacteria/isolement et purification , Humains , Mâle , Souris , Souris de lignée BALB C , Mycoplasma hyorhinis/isolement et purification , Tumeurs expérimentales/traitement médicamenteux , Tumeurs expérimentales/microbiologie , , Tumeurs du pancréas
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...