Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 547
Filter
1.
Methods Mol Biol ; 2833: 1-10, 2024.
Article in English | MEDLINE | ID: mdl-38949695

ABSTRACT

There is an increasing need for new treatment regimens to combat antibiotic-resistant strains of bacteria. Staphylococcus aureus is a clinically important, opportunist pathogen that has developed resistance to a range of antibiotics. The zebrafish larval model of systemic disease has been increasingly utilized to elucidate S. aureus virulence mechanisms and host-pathogen interactions. Here, we outline how this model can be used to investigate the effects of different antibiotics alone and in combination against S. aureus.


Subject(s)
Anti-Bacterial Agents , Disease Models, Animal , Larva , Staphylococcal Infections , Staphylococcus aureus , Zebrafish , Animals , Zebrafish/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Staphylococcus aureus/drug effects , Larva/microbiology , Larva/drug effects , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Drug Therapy, Combination , Host-Pathogen Interactions/drug effects , Microbial Sensitivity Tests
2.
Sci Rep ; 14(1): 14618, 2024 06 25.
Article in English | MEDLINE | ID: mdl-38918492

ABSTRACT

Early-life exposure to environmental toxicants like Benzo[a]pyrene (BaP) is associated with several health consequences in vertebrates (i.e., impaired or altered neurophysiological and behavioral development). Although toxicant impacts were initially studied relative to host physiology, recent studies suggest that the gut microbiome is a possible target and/or mediator of behavioral responses to chemical exposure in organisms, via the gut-brain axis. However, the connection between BaP exposure, gut microbiota, and developmental neurotoxicity remains understudied. Using a zebrafish model, we determined whether the gut microbiome influences BaP impacts on behavior development. Embryonic zebrafish were treated with increasing concentrations of BaP and allowed to grow to the larval life stage, during which they underwent behavioral testing and intestinal dissection for gut microbiome profiling via high-throughput sequencing. We found that exposure affected larval zebrafish microbiome diversity and composition in a manner tied to behavioral development: increasing concentrations of BaP were associated with increased taxonomic diversity, exposure was associated with unweighted UniFrac distance, and microbiome diversity and exposure predicted larval behavior. Further, a gnotobiotic zebrafish experiment clarified whether microbiome presence was associated with BaP exposure response and behavioral changes. We found that gut microbiome state altered the relationship between BaP exposure concentration and behavioral response. These results support the idea that the zebrafish gut microbiome is a determinant of the developmental neurotoxicity that results from chemical exposure.


Subject(s)
Behavior, Animal , Benzo(a)pyrene , Gastrointestinal Microbiome , Larva , Zebrafish , Animals , Zebrafish/microbiology , Benzo(a)pyrene/toxicity , Gastrointestinal Microbiome/drug effects , Behavior, Animal/drug effects , Larva/drug effects , Larva/microbiology
3.
Elife ; 132024 Jun 18.
Article in English | MEDLINE | ID: mdl-38896446

ABSTRACT

Tuberculosis is a major global health problem and is one of the top 10 causes of death worldwide. There is a pressing need for new treatments that circumvent emerging antibiotic resistance. Mycobacterium tuberculosis parasitises macrophages, reprogramming them to establish a niche in which to proliferate, therefore macrophage manipulation is a potential host-directed therapy if druggable molecular targets could be identified. The pseudokinase Tribbles1 (Trib1) regulates multiple innate immune processes and inflammatory profiles making it a potential drug target in infections. Trib1 controls macrophage function, cytokine production, and macrophage polarisation. Despite wide-ranging effects on leukocyte biology, data exploring the roles of Tribbles in infection in vivo are limited. Here, we identify that human Tribbles1 is expressed in monocytes and is upregulated at the transcript level after stimulation with mycobacterial antigen. To investigate the mechanistic roles of Tribbles in the host response to mycobacteria in vivo, we used a zebrafish Mycobacterium marinum (Mm) infection tuberculosis model. Zebrafish Tribbles family members were characterised and shown to have substantial mRNA and protein sequence homology to their human orthologues. trib1 overexpression was host-protective against Mm infection, reducing burden by approximately 50%. Conversely, trib1 knockdown/knockout exhibited increased infection. Mechanistically, trib1 overexpression significantly increased the levels of proinflammatory factors il-1ß and nitric oxide. The host-protective effect of trib1 was found to be dependent on the E3 ubiquitin kinase Cop1. These findings highlight the importance of Trib1 and Cop1 as immune regulators during infection in vivo and suggest that enhancing macrophage TRIB1 levels may provide a tractable therapeutic intervention to improve bacterial infection outcomes in tuberculosis.


Subject(s)
Intracellular Signaling Peptides and Proteins , Protein Serine-Threonine Kinases , Zebrafish , Animals , Humans , Disease Models, Animal , Host-Pathogen Interactions , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Macrophages/microbiology , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium Infections, Nontuberculous/genetics , Mycobacterium marinum , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Zebrafish/microbiology , Male , Female
4.
Antimicrob Agents Chemother ; 68(7): e0056124, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38899926

ABSTRACT

Staphylococcus aureus is a pathogenic bacterium responsible for a broad spectrum of infections, including cutaneous, respiratory, osteoarticular, and systemic infections. It poses a significant clinical challenge due to its ability to develop antibiotic resistance. This resistance limits therapeutic options, increases the risk of severe complications, and underscores the urgent need for new strategies to address this threat, including the investigation of treatments complementary to antibiotics. The evaluation of novel antimicrobial agents often employs animal models, with the zebrafish embryo model being particularly interesting for studying host-pathogen interactions, establishing itself as a crucial tool in this field. For the first time, this study presents a zebrafish embryo model for the in vivo assessment of bacteriophage efficacy against S. aureus infection. A localized infection was induced by microinjecting either methicillin-resistant S. aureus (MRSA) or methicillin-susceptible S. aureus (MSSA). Subsequent treatments involved administering either bacteriophage, vancomycin (the reference antibiotic for MRSA), or a combination of both via the same route to explore potential synergistic effects. Our findings indicate that the bacteriophage was as effective as vancomycin in enhancing survival rates, whether used alone or in combination. Moreover, bacteriophage treatment appears to be even more effective in reducing the bacterial load in S. aureus-infected embryos post-treatment than the antibiotic. Our study validates the use of the zebrafish embryo model and highlights its potential as a valuable tool in assessing bacteriophage efficacy treatments in vivo.


Subject(s)
Anti-Bacterial Agents , Methicillin-Resistant Staphylococcus aureus , Phage Therapy , Staphylococcal Infections , Vancomycin , Zebrafish , Animals , Zebrafish/microbiology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/virology , Phage Therapy/methods , Vancomycin/pharmacology , Vancomycin/therapeutic use , Staphylococcal Infections/therapy , Staphylococcal Infections/microbiology , Staphylococcal Infections/drug therapy , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Disease Models, Animal , Embryo, Nonmammalian/microbiology , Microbial Sensitivity Tests
5.
PLoS Biol ; 22(5): e3002606, 2024 May.
Article in English | MEDLINE | ID: mdl-38814944

ABSTRACT

Zebrafish are popular research organisms selected for laboratory use due in part to widespread availability from the pet trade. Many contemporary colonies of laboratory zebrafish are maintained in aquaculture facilities that monitor and aim to curb infections that can negatively affect colony health and confound experiments. The impact of laboratory control on the microbial constituents associated with zebrafish in research environments compared to the pet trade are unclear. Diseases of unknown causes are common in both environments. We conducted a metatranscriptomic survey to broadly compare the zebrafish-associated microbes in pet trade and laboratory environments. We detected many microbes in animals from the pet trade that were not found in laboratory animals. Cohousing experiments revealed several transmissible microbes including a newly described non-enveloped, double-stranded RNA virus in the Birnaviridae family we name Rocky Mountain birnavirus (RMBV). Infections were detected in asymptomatic animals from the pet trade, but when transmitted to laboratory animals RMBV was associated with pronounced antiviral responses and hemorrhagic disease. These experiments highlight the pet trade as a distinct source of diverse microbes that associate with zebrafish and establish a paradigm for the discovery of newly described pathogenic viruses and other infectious microbes that can be developed for study in the laboratory.


Subject(s)
Zebrafish , Animals , Zebrafish/virology , Zebrafish/microbiology , Fish Diseases/virology , Fish Diseases/microbiology , Fish Diseases/transmission , Pets/virology , Pets/microbiology , Animals, Laboratory/virology , Animals, Laboratory/microbiology , Aquaculture
6.
Microbiol Res ; 285: 127786, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38820703

ABSTRACT

The α-Gal syndrome (AGS) is an IgE-mediated tick borne-allergy that results in delayed anaphylaxis to the consumption of mammalian meat and products containing α-Gal. Considering that α-Gal-containing microbiota modulates natural antibody production to this glycan, this study aimed to evaluate the influence on tick salivary compounds on the gut microbiota composition in the zebrafish (Danio rerio) animal model. Sequencing of 16 S rDNA was performed in a total of 75 zebrafish intestine samples, representing different treatment groups: PBS control, Ixodes ricinus tick saliva, tick saliva non-protein fraction (NPF), tick saliva protein fraction (PF), and tick saliva protein fractions 1-5 with NPF (F1-5). The results revealed that treatment with tick saliva and different tick salivary fractions, combined with α-Gal-positive dog food feeding, resulted in specific variations in zebrafish gut microbiota composition at various taxonomic levels and affected commensal microbial alpha and beta diversities. Metagenomics results were corroborated by qPCR, supporting the overrepresentation of phylum Firmicutes in the tick saliva group, phylum Fusobacteriota in group F1, and phylum Cyanobacteria in F2 and F5 compared to the PBS-control. qPCRs results at genus level sustained significant enrichment of Plesiomonas spp. in groups F3 and F5, Rhizobium spp. in NPF and F4, and Cloacibacterium spp. dominance in the PBS control group. This study provides new results on the role of gut microbiota in allergic reactions to tick saliva components using a zebrafish model of AGS. Overall, gut microbiota composition in response to tick saliva biomolecules may be associated with allergic reactions to mammalian meat consumption in AGS.


Subject(s)
Food Hypersensitivity , Gastrointestinal Microbiome , Saliva , Zebrafish , Animals , Saliva/microbiology , Saliva/immunology , Zebrafish/microbiology , Food Hypersensitivity/microbiology , Food Hypersensitivity/immunology , RNA, Ribosomal, 16S/genetics , Meat , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Metagenomics , Salivary Proteins and Peptides/immunology , Salivary Proteins and Peptides/metabolism , Ixodes/microbiology , Disease Models, Animal
7.
Microbiol Res ; 285: 127770, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38788352

ABSTRACT

Edwardsiella piscicida is an acute marine pathogen that causes severe damage to the aquaculture industry worldwide. The pathogenesis of E. piscicida is dependent mainly on the type III secretion system (T3SS) and type VI secretion system (T6SS), both of which are critically regulated by EsrB and EsrC. In this study, we revealed that fatty acids influence T3SS expression. Unsaturated fatty acids (UFAs), but not saturated fatty acids (SFAs), directly interact with EsrC, which abolishes the function of EsrC and results in the turn-off of T3/T6SS. Moreover, during the in vivo colonization of E. piscicida, host fatty acids were observed to be transported into E. piscicida through FadL and to modulate the expression of T3/T6SS. Furthermore, the esrCR38G mutant blocked the interaction between EsrC and UFAs, leading to dramatic growth defects in DMEM and impaired colonization in HeLa cells and zebrafish. In conclusion, this study revealed that the interaction between UFAs and EsrC to turn off T3/T6SS expression is essential for E. piscicida infection.


Subject(s)
Bacterial Proteins , Edwardsiella , Enterobacteriaceae Infections , Fatty Acids, Unsaturated , Fish Diseases , Type III Secretion Systems , Type VI Secretion Systems , Zebrafish , Animals , Edwardsiella/genetics , Edwardsiella/metabolism , Type III Secretion Systems/metabolism , Type III Secretion Systems/genetics , Enterobacteriaceae Infections/microbiology , Humans , HeLa Cells , Zebrafish/microbiology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Type VI Secretion Systems/metabolism , Type VI Secretion Systems/genetics , Fatty Acids, Unsaturated/metabolism , Fish Diseases/microbiology , Gene Expression Regulation, Bacterial
8.
Microbiol Spectr ; 12(6): e0316823, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38722177

ABSTRACT

Vitamin B12 (B12) serves as a critical cofactor within mycobacterial metabolism. While some pathogenic strains can synthesize B12 de novo, others rely on host-acquired B12. In this investigation, we studied the transport of vitamin B12 in Mycobacterium marinum using B12-auxotrophic and B12-sensitive strains by deleting metH or metE, respectively. These two enzymes rely on B12 in different ways to function as methionine synthases. We used these strains to select mutants affecting B12 scavenging and confirmed their phenotypes during growth experiments in vitro. Our analysis of B12 uptake mechanisms revealed that membrane lipids and cell wall integrity play an essential role in cell envelope transport. Furthermore, we identified a potential transcription regulator that responds to B12. Our study demonstrates that M. marinum can take up exogenous B12 and that altering mycobacterial membrane integrity affects B12 uptake. Finally, during zebrafish infection using B12-auxotrophic and B12-sensitive strains, we found that B12 is available for virulent mycobacteria in vivo.IMPORTANCEOur study investigates how mycobacteria acquire essential vitamin B12. These microbes, including those causing tuberculosis, face challenges in nutrient uptake due to their strong outer layer. We focused on Mycobacterium marinum, similar to TB bacteria, to uncover its vitamin B12 absorption. We used modified strains unable to produce their own B12 and discovered that M. marinum can indeed absorb it from the environment, even during infections. Changes in the outer layer composition affect this process, and genes related to membrane integrity play key roles. These findings illuminate the interaction between mycobacteria and their environment, offering insights into combatting diseases like tuberculosis through innovative strategies. Our concise research underscores the pivotal role of vitamin B12 in microbial survival and its potential applications in disease control.


Subject(s)
Bacterial Outer Membrane , Mycobacterium marinum , Vitamin B 12 , Zebrafish , Mycobacterium marinum/genetics , Mycobacterium marinum/metabolism , Vitamin B 12/metabolism , Animals , Zebrafish/microbiology , Bacterial Outer Membrane/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Cell Membrane Permeability , Biological Transport , Cell Membrane/metabolism , Mycobacterium Infections, Nontuberculous/microbiology
9.
Microbiol Res ; 284: 127735, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38678681

ABSTRACT

The production of endogenous hydrogen sulfide (H2S) is an important phenotype of bacteria. H2S plays an important role in bacterial resistance to ROS and antibiotics, which significantly contributes to bacterial pathogenicity. Edwardsiella piscicida, the Gram-negative pathogen causing fish edwardsiellosis, has been documented to produce hydrogen sulfide. In the study, we revealed that Ferric uptake regulator (Fur) controlled H2S synthesis by activating the expression of phsABC operon. Besides, Fur participated in the bacterial defense against ROS and cationic antimicrobial peptides and modulated T3SS expression. Furthermore, the disruption of fur exhibited a significant in vivo colonization defect. Collectively, our study demonstrated the regulation of Fur in H2S synthesis, stress response, and virulence, providing a new perspective for better understanding the pathogenesis of Edwardsiella.


Subject(s)
Bacterial Proteins , Edwardsiella , Enterobacteriaceae Infections , Fish Diseases , Gene Expression Regulation, Bacterial , Hydrogen Sulfide , Stress, Physiological , Edwardsiella/genetics , Edwardsiella/pathogenicity , Hydrogen Sulfide/metabolism , Animals , Virulence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Enterobacteriaceae Infections/microbiology , Fish Diseases/microbiology , Repressor Proteins/metabolism , Repressor Proteins/genetics , Reactive Oxygen Species/metabolism , Operon , Antimicrobial Cationic Peptides/pharmacology , Type III Secretion Systems/metabolism , Type III Secretion Systems/genetics , Zebrafish/microbiology
10.
Development ; 151(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38577841

ABSTRACT

Enteroendocrine cells (EECs) are crucial for sensing ingested nutrients and regulating feeding behavior. How gut microbiota regulate the nutrient-sensing EEC activity is unclear. Our transcriptomic analysis demonstrates that commensal microbiota colonization significantly increases the expression of many genes associated with mitochondrial function. Using new methods to image EEC cytoplasmic and mitochondrial Ca2+ activity in live zebrafish, our data revealed that it is dynamically regulated during the EEC development process. Mature EECs display an increased mitochondrial-to-cytoplasmic Ca2+ ratio. Mitochondria are evenly distributed in the cytoplasm of immature EECs. As EECs mature, their mitochondria are highly localized at the basal membrane where EEC vesicle secretion occurs. Conventionalized (CV) EECs, but not germ-free (GF) EECs, exhibit spontaneous low-amplitude Ca2+ fluctuation. The mitochondrial-to-cytoplasmic Ca2+ ratio is significantly higher in CV EECs. Nutrient stimulants, such as fatty acid, increase cytoplasmic Ca2+ in a subset of EECs and promote a sustained mitochondrial Ca2+ and ATP increase. However, the nutrient-induced EEC mitochondrial activation is nearly abolished in GF zebrafish. Together, our study reveals that commensal microbiota are crucial in supporting EEC mitochondrial function and maturation.


Subject(s)
Calcium , Enteroendocrine Cells , Gastrointestinal Microbiome , Mitochondria , Zebrafish , Animals , Zebrafish/microbiology , Enteroendocrine Cells/metabolism , Mitochondria/metabolism , Gastrointestinal Microbiome/physiology , Calcium/metabolism , Nutrients/metabolism , Adenosine Triphosphate/metabolism
11.
Sci Rep ; 14(1): 9399, 2024 04 24.
Article in English | MEDLINE | ID: mdl-38658654

ABSTRACT

Edwardsiella piscicida causes significant economic losses to the aquaculture industry worldwide. Phage-based biocontrol methods are experiencing a renaissance because of the spread of drug-resistant genes and bacteria resulting from the heavy use of antibiotics. Here, we showed that the novel Edwardsiella phage EPP-1 could achieve comparable efficacy to florfenicol using a zebrafish model of Edwardsiella piscicida infection and could reduce the content of the floR resistance gene in zebrafish excreta. Specifically, phage EPP-1 inhibited bacterial growth in vitro and significantly improved the zebrafish survival rate in vivo (P = 0.0035), achieving an efficacy comparable to that of florfenicol (P = 0.2304). Notably, integrating the results of 16S rRNA sequencing, metagenomic sequencing, and qPCR, although the effects of phage EPP-1 converged with those of florfenicol in terms of the community composition and potential function of the zebrafish gut microbiota, it reduced the floR gene content in zebrafish excreta and aquaculture water. Overall, our study highlights the feasibility and safety of phage therapy for edwardsiellosis control, which has profound implications for the development of antibiotic alternatives to address the antibiotic crisis.


Subject(s)
Anti-Bacterial Agents , Bacteriophages , Edwardsiella , Enterobacteriaceae Infections , Thiamphenicol/analogs & derivatives , Zebrafish , Animals , Zebrafish/microbiology , Edwardsiella/genetics , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/veterinary , Enterobacteriaceae Infections/therapy , Bacteriophages/genetics , Bacteriophages/physiology , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Gastrointestinal Microbiome , Phage Therapy/methods , RNA, Ribosomal, 16S/genetics , Fish Diseases/microbiology , Fish Diseases/therapy , Fish Diseases/prevention & control , Thiamphenicol/pharmacology , Aquaculture/methods
12.
J Vis Exp ; (206)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38682933

ABSTRACT

Zebrafish serve as valuable models for research on growth, immunity, and gut microbiota due to their genomic similarities with mammals, transparent embryos developed in a relatively clean chorion environment, and extremely rapid development of larvae compared to rodent models. Germ-free (GF) zebrafish (Danio rerio) are crucial for evaluating pollutant toxicity and establishing human-like disease models related to microbial functions. In comparison to conventionally raised (CR) models (fish in common husbandry), GF zebrafish allow for more accurate manipulation of the host microbiota, aiding in determining the causal relationship between microorganisms and hosts. Consequently, they play a critical role in advancing our understanding of these relationships. However, GF zebrafish models are typically generated and researched during the early life stages (from embryos to larvae) due to limitations in immune function and nutrient absorption. This study optimizes the generation, maintenance, and identification of early GF zebrafish models without feeding and with long-term feeding using GF food (such as Artemia sp., brine shrimp). Throughout the process, daily sampling and culture were performed and identified through multiple detections, including plates and 16S rRNA sequencing. The aseptic rate, survival, and developmental indexes of GF zebrafish were recorded to ensure the quality and quantity of the generated models. Importantly, this study provides details on bacterial isolation and infection techniques for GF fish, enabling the efficient creation of GF fish models from larvae to juvenile stages with GF food support. By applying these procedures in biomedical research, scientists can better understand the relationships between intestinal bacterial functions and host health.


Subject(s)
Germ-Free Life , Larva , Models, Animal , Zebrafish , Animals , Zebrafish/microbiology , Larva/microbiology , Larva/growth & development , Female , Male
13.
Physiol Genomics ; 56(6): 426-435, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38557279

ABSTRACT

Short-chain fatty acids (SCFAs) produced by the gut bacteria have been associated with cardiovascular dysfunction in humans and rodents. However, studies exploring effects of SCFAs on cardiovascular parameters in the zebrafish, an increasingly popular model in cardiovascular research, remain limited. Here, we performed fecal bacterial 16S sequencing and gas chromatography/mass spectrometry (GC-MS) to determine the composition and abundance of gut microbiota and SCFAs in adult zebrafish. Following this, the acute effects of major SCFAs on heart rate and vascular tone were measured in anesthetized zebrafish larvae using fecal concentrations of butyrate, acetate, and propionate. Finally, we investigated if coincubation with butyrate may lessen the effects of angiotensin II (ANG II) and phenylephrine (PE) on vascular tone in anesthetized zebrafish larvae. We found that the abundance in Proteobacteria, Firmicutes, and Fusobacteria phyla in the adult zebrafish resembled those reported in rodents and humans. SCFA levels with highest concentration of acetate (27.43 µM), followed by butyrate (2.19 µM) and propionate (1.65 µM) were observed in the fecal samples of adult zebrafish. Immersion in butyrate and acetate produced a ∼20% decrease in heart rate (HR), respectively, with no observed effects of propionate. Butyrate alone also produced an ∼25% decrease in the cross-sectional width of the dorsal aorta (DA) at 60 min (*P < 0.05), suggesting compensatory vasoconstriction, with no effects of either acetate or propionate. In addition, butyrate significantly alleviated the decrease in DA cross-sectional width produced by both ANG II and PE. We demonstrate the potential for zebrafish in investigation of host-microbiota interactions in cardiovascular health.NEW & NOTEWORTHY We highlight the presence of a core gut microbiota and demonstrate in vivo short-chain fatty acid production in adult zebrafish. In addition, we show cardio-beneficial vasoactive and chronotropic properties of butyrate, and chronotropic properties of acetate in anesthetized zebrafish larvae.


Subject(s)
Fatty Acids, Volatile , Feces , Gastrointestinal Microbiome , Heart Rate , Larva , Zebrafish , Animals , Zebrafish/microbiology , Gastrointestinal Microbiome/drug effects , Fatty Acids, Volatile/metabolism , Heart Rate/drug effects , Feces/microbiology , Butyrates/metabolism , Butyrates/pharmacology , Angiotensin II/metabolism , Angiotensin II/pharmacology , Bacteria/drug effects , Phenylephrine/pharmacology , Acetates/pharmacology , Acetates/metabolism , RNA, Ribosomal, 16S/genetics
14.
mBio ; 15(6): e0034624, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38682911

ABSTRACT

Staphylococcus aureus is a major pathogen associated with important diseases in humans and animals. Macrophages are a key component of the innate immune response to S. aureus infection and play a major role in disease outcomes. To investigate the adaptive evolution of S. aureus in response to macrophages, we developed an experimental infection assay. S. aureus strains representing major human epidemic clones were passaged many times in a macrophage cell line, accumulating mutations in an array of genomic loci. Phenotypic analysis revealed the emergence of a lineage exhibiting increased survival in macrophages and human blood, and resistance to vancomycin. The evolved lineage exhibited a previously undescribed small colony variant (SCV) phenotype characterized by hyper-pigmentation, which resulted from a missense mutation in rsbW. Notably, the novel SCV was a conditional adaptive trait that was unstable in nutrient-replete conditions in vitro, rapidly converting from hyper-pigmented SCV to a non-pigmented large colony variant via spontaneous sigB deletion events. Importantly, we identified similar deletions in the genome sequences of a limited number of clinical S. aureus isolates from public databases, indicating that related events may occur during clinical infection. Experimental infection of zebrafish did not reveal a difference in virulence between parent and novel SCV but demonstrated an in vivo fitness cost for the compensatory sigB deletion events. Taken together, we report an experimental evolutionary approach for investigating bacterial innate immune cell interactions, revealing a conditional adaptation that promotes S. aureus survival in macrophages and resistance to vancomycin. IMPORTANCE: Staphylococcus aureus is an important human bacterial pathogen. The host response to S. aureus involves the production of innate immune cells such as macrophages which are important for fighting infection. Here we report a new model of experimental evolution for studying how S. aureus can evade killing by macrophages. We identified a novel adaptive phenotype that promotes survival in macrophages and blood and resistance to antibiotics. The phenotype is lost rapidly upon growth in nutrient-rich conditions via disruption of the alternative sigma factor sigB, revealing a conditional niche-specific fitness advantage. Genomic analysis of clinical isolates suggests similar adaptations may occur during human infections. Our model may be used broadly to identify adaptations of S. aureus to the innate immune response.


Subject(s)
Macrophages , Staphylococcal Infections , Staphylococcus aureus , Zebrafish , Staphylococcus aureus/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Macrophages/microbiology , Macrophages/immunology , Humans , Animals , Staphylococcal Infections/microbiology , Staphylococcal Infections/immunology , Zebrafish/microbiology , Phenotype , Microbial Viability , Anti-Bacterial Agents/pharmacology , Adaptation, Physiological/genetics , Cell Line , Mice , Genome, Bacterial , Evolution, Molecular
15.
J Fish Biol ; 104(6): 2068-2080, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38596840

ABSTRACT

The pervasive utilization of plastics and their integration into ecosystems has resulted in significant environmental issues, particularly the pollution of microplastics (MPs). In aquaculture, high-fat feed (HFD) is frequently employed to enhance the energy intake and economic fish production. This study utilized zebrafish as a model organism to investigate the impact of concurrent exposure to HFD and MPs on fish intestinal pathology damage and intestinal microbiome. The experimental design involved the division of zebrafish into two groups: one receiving a normal diet (ND) and the other receiving HFD. The zebrafish were exposed to a control group, as well as polystyrene (PS) MPs of varying sizes (5 and 50 µm). Histopathological examination revealed that the combination of 5 µm MPs and HFD resulted in the most significant damage to the zebrafish intestinal tract. Furthermore, gut microbiome assays indicated that exposure to MPs and HFD altered the composition of the gut microbiome. This study demonstrates that in aquaculture, the issue of HFD must be considered alongside concerns about MPs contamination, as both factors appear to have a combined effect on the intestinal pathology damage and intestinal microbiome. The findings of this research offer valuable insights for the improvement of fish farming practices.


Subject(s)
Gastrointestinal Microbiome , Intestines , Microplastics , Polystyrenes , Water Pollutants, Chemical , Zebrafish , Animals , Zebrafish/microbiology , Microplastics/toxicity , Polystyrenes/toxicity , Polystyrenes/adverse effects , Gastrointestinal Microbiome/drug effects , Intestines/pathology , Intestines/microbiology , Intestines/drug effects , Water Pollutants, Chemical/toxicity , Water Pollutants, Chemical/adverse effects , Aquaculture , Diet, High-Fat/adverse effects , Animal Feed/analysis
16.
Int. microbiol ; 27(1): 25-35, Feb. 2024. graf, ilus
Article in English | IBECS | ID: ibc-230241

ABSTRACT

Pseudomonas is a group of bacteria that can cause a wide range of infections, particularly in people with weakened immune systems, such as those with cystic fibrosis or who are hospitalized. It can also cause infections in the skin and soft tissue, including cellulitis, abscesses and wound infections. Antimicrobial peptides (AMPS) are the alternative strategy due to their broad spectrum of activity and act as effective treatment against multi-drug resistance pathogens. In this study, we have used an AMP, RW20 (1RPVKRKKGWPKGVKRGPPKW20). RW20 peptide is derived from the histone acetyltransferases (HATs) of the freshwater teleost, Channa striatus. The antimicrobial prediction tool has been utilized to identify the RW20 sequence from the HATs sequence. We synthesized the peptide to explore its mechanism of action. In an in vitro assay, RW20 was challenged against P. aeruginosa and we showed that RW20 displayed antibacterial properties and damaged the cell membrane. The mechanism of action of RW20 against P. aeruginosa has been established via field emission scanning electron microscopy (FESEM) as well as fluorescence assisted cell sorter (FACS) analysis. Both these experiments established that RW20 caused bacterial membrane disruption and cell death. Moreover, the impact of RW20, in-vivo, was tested against P. aeruginosa-infected zebrafish larvae. In the infected larvae, RW20 showed protective effect against P. aeruginosa by increasing the larval antioxidant enzymes, reducing the excess oxidative stress and apoptosis. Thus, it is possible that HATs-derived RW20 can be an efficient antimicrobial molecule against P. aeruginosa.(AU)


Subject(s)
Humans , Histone Acetyltransferases/administration & dosage , Pseudomonas aeruginosa , Zebrafish/microbiology , Larva , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/metabolism , Microbiology , Microbiological Techniques , Microbial Sensitivity Tests , Pseudomonas Infections
17.
Infect Immun ; 92(2): e0038023, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38168666

ABSTRACT

Macrophages act as a first line of defense against pathogens. Against Aspergillus fumigatus, a fungus with pathogenic potential in immunocompromised patients, macrophages can phagocytose fungal spores and inhibit spore germination to prevent the development of tissue-invasive hyphae. However, the cellular pathways that macrophages use to accomplish these tasks and any roles macrophages have later in infection against invasive forms of fungi are still not fully known. Rac-family Rho GTPases are signaling hubs for multiple cellular functions in leukocytes, including cell migration, phagocytosis, reactive oxygen species (ROS) generation, and transcriptional activation. We therefore aimed to further characterize the function of macrophages against A. fumigatus in an in vivo vertebrate infection model by live imaging of the macrophage behavior in A. fumigatus-infected rac2 mutant zebrafish larvae. While Rac2-deficient zebrafish larvae are susceptible to A. fumigatus infection, Rac2 deficiency does not impair macrophage migration to the infection site, interaction with and phagocytosis of spores, spore trafficking to acidified compartments, or spore killing. However, we reveal a role for Rac2 in macrophage-mediated inhibition of spore germination and control of invasive hyphae. Re-expression of Rac2 under a macrophage-specific promoter rescues the survival of A. fumigatus-infected rac2 mutant larvae through increased control of germination and hyphal growth. Altogether, we describe a new role for macrophages against extracellular hyphal growth of A. fumigatus and report that the function of the Rac2 Rho GTPase in macrophages is required for this function.


Subject(s)
Aspergillosis , Zebrafish , Animals , Humans , Zebrafish/microbiology , GTP Phosphohydrolases , Macrophages/microbiology , Phagocytosis , Aspergillosis/microbiology , Aspergillus fumigatus/physiology , Spores, Fungal , rac GTP-Binding Proteins/genetics , Zebrafish Proteins/genetics
18.
J Antibiot (Tokyo) ; 77(4): 245-256, 2024 04.
Article in English | MEDLINE | ID: mdl-38238588

ABSTRACT

Tunicamycins (TUN) are well-defined, Streptomyces-derived natural products that inhibit protein N-glycosylation in eukaryotes, and by a conserved mechanism also block bacterial cell wall biosynthesis. TUN inhibits the polyprenylphosphate-N-acetyl-hexosamine-1-phospho-transferases (PNPT), an essential family of enzymes found in both bacteria and eukaryotes. We have previously published the development of chemically modified TUN, called TunR1 and TunR2, that have considerably reduced activity on eukaryotes but that retain the potent antibacterial properties. A mechanism for this reduced toxicity has also been reported. TunR1 and TunR2 have been tested against mammalian cell lines in culture and against live insect cells but, until now, no in vivo evaluation has been undertaken for vertebrates. In the current work, TUN, TunR1, and TunR2 are investigated for their relative toxicity and antimycobacterial activity in zebrafish using a well-established Mycobacterium marinum (M. marinum) infection system, a model for studying human Mycobacterium tuberculosis infections. We also report the relative ability to activate the unfolded protein response (UPR), the known mechanism for the eukaryotic toxicity observed with TUN treatment. Importantly, TunR1 and TunR2 retained their antimicrobial properties, as evidenced by a reduction in M. marinum bacterial burden, compared to DMSO-treated zebrafish. In summary, findings from this study highlight the characteristics of recently developed TUN derivatives, mainly TunR2, and its potential for use as a novel anti-bacterial agent for veterinary and potential medical purposes.


Subject(s)
Mycobacterium Infections, Nontuberculous , Mycobacterium marinum , Tunicamycin , Animals , Humans , Anti-Bacterial Agents/pharmacology , Mammals , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium marinum/physiology , Tunicamycin/chemistry , Tunicamycin/analogs & derivatives , Zebrafish/microbiology , Phosphotransferases/chemistry
19.
mSphere ; 9(1): e0050423, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38073033

ABSTRACT

Cryptococcus is a ubiquitous environmental fungus and frequent colonizer of human lungs. Colonization can lead to diverse outcomes, from clearance to long-term colonization to life-threatening meningoencephalitis. Regardless of the outcome, the process starts with an encounter with phagocytes. Using the zebrafish model of this infection, we have noted that cryptococcal cells first spend time inside macrophages before they become capable of pathogenic replication and dissemination. What "licensing" process takes place during this initial encounter, and how are licensed cryptococcal cells different? To address this, we isolated cryptococcal cells after phagocytosis by cultured macrophages and found these macrophage-experienced cells to be markedly more virulent in both zebrafish and mouse models. Despite producing a thick polysaccharide capsule, they were still subject to phagocytosis by macrophages in the zebrafish. Analysis of antigenic cell wall components in these licensed cells demonstrated that components of mannose and chitin are more available for staining than they are in culture-grown cells or cells with capsule production induced in vitro. Cryptococcus is capable of exiting or transferring between macrophages in vitro, raising the likelihood that this fungus alternates between intracellular and extracellular life during growth in the lungs. Our results raise the possibility that intracellular life has its advantages over time, and phagocytosis-induced alteration in mannose and chitin exposure is one way that makes subsequent rounds of phagocytosis more beneficial to the fungus.IMPORTANCECryptococcosis begins in the lungs and can ultimately travel through the bloodstream to cause devastating infection in the central nervous system. In the zebrafish model, small amounts of cryptococcus inoculated into the bloodstream are initially phagocytosed and become far more capable of dissemination after they exit macrophages. Similarly, survival in the mouse lung produces cryptococcal cell types with enhanced dissemination. In this study, we have evaluated how phagocytosis changes the properties of Cryptococcus during pathogenesis. Macrophage-experienced cells (MECs) become "licensed" for enhanced virulence. They out-disseminate culture-grown cells in the fish and out-compete non-MECs in the mouse lung. Analysis of their cell surface demonstrates that MECs have increased availability of cell wall components mannose and chitin substances involved in provoking phagocytosis. These findings suggest how Cryptococcus might tune its cell surface to induce but survive repeated phagocytosis during early pathogenesis in the lung.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Animals , Mice , Humans , Zebrafish/microbiology , Cryptococcosis/microbiology , Virulence , Mannose , Macrophages/microbiology , Chitin/metabolism
20.
mBio ; 15(1): e0225923, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38063379

ABSTRACT

IMPORTANCE: Phase variation allows a single strain to produce phenotypic diverse subpopulations. Phase-variable restriction modification (RM) systems are systems that allow for such phase variation via epigenetic regulation of gene expression levels. The phase-variable RM system SsuCC20p was found in multiple streptococcal species and was acquired by an emerging zoonotic lineage of Streptococcus suis. We show that the phase variability of SsuCC20p is dependent on a recombinase encoded within the SsuCC20p locus. We characterized the genome methylation profiles of the different phases of SsuCC20p and demonstrated the consequential impact on the transcriptome and virulence in a zebrafish infection model. Acquiring mobile genetic elements containing epigenetic regulatory systems, like phase-variable RM systems, enables bacterial pathogens to produce diverse phenotypic subpopulations that are better adapted to specific (host) environments encountered during infection.


Subject(s)
Streptococcal Infections , Streptococcus suis , Animals , Streptococcus suis/genetics , Streptococcus suis/metabolism , Epigenesis, Genetic , DNA Restriction-Modification Enzymes/genetics , Zebrafish/microbiology , Virulence , Larva/microbiology , Epigenome , Transcriptome , Streptococcal Infections/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL