Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 532
Filtrar
1.
Sci Rep ; 14(1): 9399, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38658654

RESUMO

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.


Assuntos
Antibacterianos , Bacteriófagos , Edwardsiella , Infecções por Enterobacteriaceae , Tianfenicol/análogos & derivados , Peixe-Zebra , Animais , Peixe-Zebra/microbiologia , Edwardsiella/genética , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/veterinária , Infecções por Enterobacteriaceae/terapia , Bacteriófagos/genética , Bacteriófagos/fisiologia , Antibacterianos/farmacologia , Farmacorresistência Bacteriana/genética , Microbioma Gastrointestinal , Terapia por Fagos/métodos , RNA Ribossômico 16S/genética , Doenças dos Peixes/microbiologia , Doenças dos Peixes/terapia , Doenças dos Peixes/prevenção & controle , Tianfenicol/farmacologia , Aquicultura/métodos
2.
Int. microbiol ; 27(1): 25-35, Feb. 2024. graf, ilus
Artigo em Inglês | IBECS | ID: ibc-230241

RESUMO

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)


Assuntos
Humanos , Histona Acetiltransferases/administração & dosagem , Pseudomonas aeruginosa , Peixe-Zebra/microbiologia , Larva , Antibacterianos/farmacologia , Anti-Infecciosos/metabolismo , Microbiologia , Técnicas Microbiológicas , Testes de Sensibilidade Microbiana , Infecções por Pseudomonas
3.
Infect Immun ; 92(2): e0038023, 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38168666

RESUMO

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.


Assuntos
Aspergilose , Peixe-Zebra , Animais , Humanos , Peixe-Zebra/microbiologia , GTP Fosfo-Hidrolases , Macrófagos/microbiologia , Fagocitose , Aspergilose/microbiologia , Aspergillus fumigatus/fisiologia , Esporos Fúngicos , Proteínas rac de Ligação ao GTP/genética , Proteínas de Peixe-Zebra/genética
4.
J Antibiot (Tokyo) ; 77(4): 245-256, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38238588

RESUMO

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.


Assuntos
Infecções por Mycobacterium não Tuberculosas , Mycobacterium marinum , Tunicamicina , Animais , Humanos , Antibacterianos/farmacologia , Mamíferos , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium marinum/fisiologia , Tunicamicina/química , Tunicamicina/análogos & derivados , Peixe-Zebra/microbiologia , Fosfotransferases/química
5.
mBio ; 15(1): e0225923, 2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38063379

RESUMO

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.


Assuntos
Infecções Estreptocócicas , Streptococcus suis , Animais , Streptococcus suis/genética , Streptococcus suis/metabolismo , Epigênese Genética , Enzimas de Restrição-Modificação do DNA/genética , Peixe-Zebra/microbiologia , Virulência , Larva/microbiologia , Epigenoma , Transcriptoma , Infecções Estreptocócicas/microbiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo
6.
J Leukoc Biol ; 115(3): 525-535, 2024 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-37982587

RESUMO

Because granulomas are a hallmark of tuberculosis pathogenesis, the study of the dynamic changes in their cellular composition and morphological character can facilitate our understanding of tuberculosis pathogenicity. Adult zebrafish infected with Mycobacterium marinum form granulomas that are similar to the granulomas in human patients with tuberculosis and therefore have been used to study host-mycobacterium interactions. Most studies of zebrafish granulomas, however, have focused on necrotic granulomas, while a systematic description of the different stages of granuloma formation in the zebrafish model is lacking. Here, we characterized the stages of granulomas in M. marinum-infected zebrafish, including early immune cell infiltration, nonnecrotizing granulomas, and necrotizing granulomas, using corresponding samples from patients with pulmonary tuberculosis as references. We combined hematoxylin and eosin staining and in situ hybridization to identify the different immune cell types and follow their spatial distribution in the different stages of granuloma development. The macrophages in zebrafish granulomas were shown to belong to distinct subtypes: epithelioid macrophages, foamy macrophages, and multinucleated giant cells. By defining the developmental stages of zebrafish granulomas and the spatial distribution of the different immune cells they contain, this work provides a reference for future studies of mycobacterial granulomas and their immune microenvironments.


Assuntos
Infecções por Mycobacterium não Tuberculosas , Mycobacterium tuberculosis , Mycobacterium , Tuberculose , Animais , Humanos , Peixe-Zebra/microbiologia , Granuloma/microbiologia , Granuloma/patologia
7.
mSphere ; 9(1): e0050423, 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38073033

RESUMO

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.


Assuntos
Criptococose , Cryptococcus neoformans , Animais , Camundongos , Humanos , Peixe-Zebra/microbiologia , Criptococose/microbiologia , Virulência , Manose , Macrófagos/microbiologia , Quitina/metabolismo
8.
Sci Total Environ ; 912: 169303, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38135076

RESUMO

A plethora of studies have so far described the toxic effects of bisphenol A (BPA) on organism health, highlighting the urgent need to find new strategies not only to reduce the presence of this toxicant but also to counteract its adverse effects. In this context, probiotics emerged as a potential tool since they promote organism welfare. Using a multidisciplinary approach, this study explores the effects of SLAB51 dietary administration to counteract BPA toxicity using zebrafish as a model. Adult males and females were maintained under standard conditions (control group; C), exposed for 28 days via the water to an environmental relevant dose of BPA (10 µg/L; BPA), dietary treated with SLAB51 (109 CFU/g of body weight; P) and co-treated with BPA plus SLAB51 (BPA + P). In the gut, exposure to BPA resulted in altered architecture in both males and females, with females also experiencing an increase of pathogenic bacterial species. Co-administration of BPA + P led to the restoration of normal gut architecture, favored beneficial bacteria colonization, and decreased the abundance of pathogenic species. In the liver, male BPA exposure led to steatosis and glycogen depletion, which was partially mitigated by SLAB51 co-administration. In contrast, in females exposed to BPA, the lack of steatosis along with the greater glycogen depletion, suggested an increase in energy demand as supported by the metabolomic phenotype. The analysis of liver metabolites in BPA + P males revealed increased levels of anserine and reduced levels of glutamine, which could lie behind the counteraction of the brain histopathological damage caused by BPA. In BPA + P females, a reduction of retinoic acid was found in the liver, suggesting an increase in retinoids responsible for BPA detoxification. Overall, these results demonstrate that SLAB51 exerts its beneficial effects on the gut microbiota-brain-liver axis through distinct molecular pathways, effectively mitigating the pleiotropic toxicity of BPA.


Assuntos
Disruptores Endócrinos , Fígado Gorduroso , Microbioma Gastrointestinal , Fenóis , Probióticos , Animais , Feminino , Masculino , Peixe-Zebra/microbiologia , Compostos Benzidrílicos/toxicidade , Encéfalo , Glicogênio , Disruptores Endócrinos/toxicidade
9.
Dis Model Mech ; 17(1)2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38131137

RESUMO

Shigella flexneri is a human-adapted pathovar of Escherichia coli that can invade the intestinal epithelium, causing inflammation and bacillary dysentery. Although an important human pathogen, the host response to S. flexneri has not been fully described. Zebrafish larvae represent a valuable model for studying human infections in vivo. Here, we use a Shigella-zebrafish infection model to generate mRNA expression profiles of host response to Shigella infection at the whole-animal level. Immune response-related processes dominate the signature of early Shigella infection (6 h post-infection). Consistent with its clearance from the host, the signature of late Shigella infection (24 h post-infection) is significantly changed, and only a small set of immune-related genes remain differentially expressed, including acod1 and gpr84. Using mutant lines generated by ENU, CRISPR mutagenesis and F0 crispants, we show that acod1- and gpr84-deficient larvae are more susceptible to Shigella infection. Together, these results highlight the power of zebrafish to model infection by bacterial pathogens and reveal the mRNA expression of the early (acutely infected) and late (clearing) host response to Shigella infection.


Assuntos
Disenteria Bacilar , Animais , Humanos , Disenteria Bacilar/genética , Shigella flexneri/genética , Shigella flexneri/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/microbiologia , Inflamação/microbiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
10.
Int J Mol Sci ; 24(24)2023 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-38139043

RESUMO

Interleukin-6 (IL-6), a pleiotropic cytokine, plays a crucial role in acute stress induced by bacterial infection and is strongly associated with reactive oxygen species (ROS) production. However, the role of IL-6 in the liver of fish after Aeromonas hydrophila infection remains unclear. Therefore, this study constructed a zebrafish (Danio rerio) il-6 knockout line by CRISPR/Cas9 to investigate the function of IL-6 in the liver post bacterial infection. After infection with A. hydrophila, pathological observation showed that il-6-/- zebrafish exhibited milder liver damage than wild-type (WT) zebrafish. Moreover, liver transcriptome sequencing revealed that 2432 genes were significantly up-regulated and 1706 genes were significantly down-regulated in il-6-/- fish compared with WT fish after A. hydrophila infection. Further, gene ontology (GO) analysis showed that differentially expressed genes (DEGs) were significantly enriched in redox-related terms, including oxidoreductase activity, copper ion transport, etc. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that DEGs were significantly enriched in pathways such as the PPAR signaling pathway, suggesting that il-6 mutation has a significant effect on redox processes in the liver after A. hydrophila infection. Additionally, il-6-/- zebrafish exhibited lower malondialdehyde (MDA) levels and higher superoxide dismutase (SOD) activities in the liver compared with WT zebrafish following A. hydrophila infection, indicating that IL-6 deficiency mitigates oxidative stress induced by A. hydrophila infection in the liver. These findings provide a basis for further studies on the role of IL-6 in regulating oxidative stress in response to bacterial infections.


Assuntos
Infecções Bacterianas , Infecções por Bactérias Gram-Negativas , Interleucina-6 , Proteínas de Peixe-Zebra , Animais , Aeromonas hydrophila/fisiologia , Infecções Bacterianas/patologia , Infecções por Bactérias Gram-Negativas/genética , Infecções por Bactérias Gram-Negativas/microbiologia , Interleucina-6/genética , Fígado/patologia , Mutação , Oxirredução , Estresse Oxidativo/genética , Peixe-Zebra/genética , Peixe-Zebra/microbiologia , Proteínas de Peixe-Zebra/metabolismo , Modelos Animais de Doenças
11.
Cell Rep ; 42(11): 113407, 2023 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-37948182

RESUMO

Vibrio cholerae is an aquatic bacterium that causes severe and potentially deadly diarrheal disease. Despite the impact on global health, our understanding of host mucosal responses to Vibrio remains limited, highlighting a knowledge gap critical for the development of effective prevention and treatment strategies. Using a natural infection model, we combine physiological and single-cell transcriptomic studies to characterize conventionally reared adult zebrafish guts and guts challenged with Vibrio. We demonstrate that Vibrio causes a mild mucosal immune response characterized by T cell activation and enhanced antigen capture; Vibrio suppresses host interferon signaling; and ectopic activation of interferon alters the course of infection. We show that the adult zebrafish gut shares similarities with mammalian counterparts, including the presence of Best4+ cells, tuft cells, and a population of basal cycling cells. These findings provide important insights into host-pathogen interactions and emphasize the utility of zebrafish as a natural model of Vibrio infection.


Assuntos
Cólera , Vibrio cholerae , Animais , Cólera/microbiologia , Peixe-Zebra/microbiologia , Intestinos/microbiologia , Interferons , Mamíferos
12.
Microbiome ; 11(1): 252, 2023 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-37951983

RESUMO

BACKGROUND: Perturbations of animal-associated microbiomes from chemical stress can affect host physiology and health. While dysbiosis induced by antibiotic treatments and disease is well known, chemical, nonantibiotic drugs have recently been shown to induce changes in microbiome composition, warranting further exploration. Loperamide is an opioid-receptor agonist widely prescribed for treating acute diarrhea in humans. Loperamide is also used as a tool to study the impact of bowel dysfunction in animal models by inducing constipation, but its effect on host-associated microbiota is poorly characterized. RESULTS: We used conventional and gnotobiotic larval zebrafish models to show that in addition to host-specific effects, loperamide also has anti-bacterial activities that directly induce changes in microbiota diversity. This dysbiosis is due to changes in bacterial colonization, since gnotobiotic zebrafish mono-colonized with bacterial strains sensitive to loperamide are colonized up to 100-fold lower when treated with loperamide. Consistently, the bacterial diversity of gnotobiotic zebrafish colonized by a mix of 5 representative bacterial strains is affected by loperamide treatment. CONCLUSION: Our results demonstrate that loperamide, in addition to host effects, also induces dysbiosis in a vertebrate model, highlighting that established treatments can have underlooked secondary effects on microbiota structure and function. This study further provides insights for future studies exploring how common medications directly induce changes in host-associated microbiota. Video Abstract.


Assuntos
Loperamida , Microbiota , Humanos , Animais , Loperamida/efeitos adversos , Peixe-Zebra/microbiologia , Disbiose/induzido quimicamente , Constipação Intestinal/induzido quimicamente , Bactérias
13.
Front Cell Infect Microbiol ; 13: 1271448, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37868352

RESUMO

Background: Aeromonas hydrophila is an important pathogen that mainly harms aquatic animals and exhibits resistance to a variety of antibiotics. This study investigated the effect of epigallocatechin-3-gallate (EGCG) on the virulence factors of A.hydrophila and its impact on adhesion, invasion, and cytotoxicity in Caco-2 cells. The potential mechanism of antibacterial activity of EGCG was investigated by transcriptomic analysis. Results: EGCG not only inhibited the production of biofilm, hemolytic activity, motility, and protease activity of A.hydrophila, but also reduced its adhesion, invasion, and cytotoxicity in Caco-2 cells. Transcriptomic analysis indicated that the antimicrobial activity of EGCG may be achieved by weakening the chemotaxis and stress response of the bacteria, as well as inhibiting the TonB system. Animal studies demonstrated that EGCG can significantly improve the survival rate and organs damage of zebrafish infected with A.hydrophila. Conclusion: EGCG would be a potential alternative drug for the prevention and treatment of A. hydrophila infections by anti-virulence mechanism.


Assuntos
Aeromonas hydrophila , Infecções por Bactérias Gram-Negativas , Animais , Humanos , Aeromonas hydrophila/genética , Peixe-Zebra/microbiologia , Células CACO-2 , Transcriptoma , Antibacterianos/farmacologia , Infecções por Bactérias Gram-Negativas/tratamento farmacológico , Infecções por Bactérias Gram-Negativas/microbiologia
14.
Int. microbiol ; 26(3): 551-561, Ene-Agos, 2023. graf
Artigo em Inglês | IBECS | ID: ibc-223981

RESUMO

The gut-brain axis directly regulates the brain homeostatic environment; an imbalance in gut microbial composition following ethanol exposure is maleficent. In this context, involvement of probiotics as prophylactic intervention against ethanol-induced neurotoxicity is elusive in the literature. Therefore, the present study was aimed to determine the impact of chronic ethanol exposure on the neurobehavioral response of zebrafish and possible neuroprotection through co-supplementation of probiotic Lactobacillus rhamnosus GG (LGG). Zebrafish were divided into naive, control, ethanol (0.01% v/v), LGG, and ethanol co-supplemented with LGG groups. Neurobehavioral assessment was performed after 7 days of chronic waterborne exposure to ethanol with LGG co-supplementation followed by histopathological studies. The findings indicated that there was a clear alteration in locomotor activity and habitat preference, with animals preferentially migrating toward altered zones on exposure to ethanol. However, co-supplementation of LGG showed restoration against ethanol-induced neurobehavioral and cognitive dysfunction. Brain tissue pyknosis and intestinal epithelial disruption were significantly mitigated on LGG co-supplementation against ethanol in zebrafish. The present study provides a novel approach toward supplementation of probiotics such as LGG in modulation of gut commensal microbiota influencing zebrafish behavior. Moreover, the findings delineate the possible role of probiotics as a curative administration to counter ethanol-persuaded neurological outcomes.(AU)


Assuntos
Humanos , Lacticaseibacillus rhamnosus , Peixe-Zebra/microbiologia , Etanol , Mucosa Intestinal , Microbioma Gastrointestinal , Microbiologia , Técnicas Microbiológicas
15.
Methods Mol Biol ; 2674: 313-326, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37258977

RESUMO

Mycobacterial infections, including tuberculosis, are a major health problem globally. Prevention and treatments of tuberculosis are challenging due to the poor efficacy of the current vaccine and the emergence of drug-resistant strains. Therefore, it is critical to increase our basic understanding of mycobacterial virulence strategies as well as the host immune response during infection in the complex in vivo setting. While existing infection models provide valuable tools for investigating mycobacterial pathogenesis, they also exhibit limitations that can be addressed by the development of complementary models. Here we describe recent advances to the murine Mycobacterium marinum infection model, in which the bacteria produce a local infection restricted to the tail tissue. The M. marinum model has the advantage of mimicking some of the key hallmarks of human tuberculosis not replicated in the conventional murine Mycobacterium tuberculosis model, such as the formation of granulomas with central caseating necrosis and the spontaneous development of a latency-like stage. Moreover, the model is non-lethal and enables longitudinal analysis of disease development in live animals. In this chapter, we report protocols to prepare infected tissue samples for detailed and quantitative analysis of the immune response by flow cytometry, immunofluorescence microscopy, RT-qPCR, ELISA, and Western blot, as well as for the analysis of bacterial load and localization.


Assuntos
Infecções por Mycobacterium não Tuberculosas , Mycobacterium marinum , Mycobacterium tuberculosis , Tuberculose , Animais , Camundongos , Humanos , Infecções por Mycobacterium não Tuberculosas/microbiologia , Tuberculose/microbiologia , Virulência , Peixe-Zebra/microbiologia
16.
Curr Microbiol ; 80(5): 136, 2023 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-36914801

RESUMO

It is known that probiotic microorganisms play important roles in the composition of the intestinal microbiota. Also, probiotics can affect the paracellular and transcellular transport mechanisms performed by intestinal cells. The aim of this work was to evaluate the effect of the potential probiotic Bacillus subtilis KM0 on the profile of the gut microbiota and transcription of genes related to intestinal transport of zebrafish (Danio rerio). Zebrafish was exposed by immersion to B. subtilis KM0 for 48 h, and the intestines were collected for metataxonomic analysis and transcription of genes related to transcellular and paracellular transports. Although exposure to B. subtilis changed the intestinal microbiota profile of zebrafish, the diversity indices were not altered. A decrease in the number of genera of potentially pathogenic bacteria (Flavobacterium, Plesiomonas, and Pseudomonas) and downregulation in transcription of transcellular transport genes (cubn and amn) were observed. B. subtilis KM0 strain had the expected probiotic effect, by interfering with the proliferation of potentially pathogenic bacteria and decreasing the transcription of genes codifying for signals involved with a mechanism that can be used for invasion by pathogens. The present study demonstrated that, even with a short-term exposure, a bacterium with probiotic potential such as the KM0 strain of B. subtilis can modify the profile of the host's intestinal microbiota, with an impact on the regulation of intestinal genes related to mechanisms that can be used for invasion by pathogenic bacteria.


Assuntos
Microbioma Gastrointestinal , Probióticos , Animais , Bacillus subtilis/genética , Peixe-Zebra/microbiologia , Intestinos/microbiologia
17.
Aquat Toxicol ; 257: 106459, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36857871

RESUMO

With large amounts of cephalosporin end up in natural ecosystems, water has been acknowledged as the large reservoir of ß-lactam resistance over the past decades. However, there is still insufficient knowledge available on the function of the living organisms to the transmission of antibiotic resistance. For this reason, in this study, using adult zebrafish (Danio rerio) as animal model, exposing them to environmentally relevant dose of cefotaxime for 150 days, we asked whether cefotaxime contamination accelerated ß-lactam resistance in gut microbiota as well as its potential transmission. Results showed that some of ß-lactam resistance genes (ßRGs) were intrinsic embedded in intestinal microbiome of zebrafish even without antibiotic stressor. Across cefotaxime treatment, the abundance of most ßRGs in fish gut microbiome decreased apparently in the short term firstly, and then increased with the prolonged exposure, forming distinctly divergent ßRG profiles with antibiotic-untreated zebrafish. Meanwhile, with the rising concentration of cefotaxime, the range of ßRGs' host-taxa expanded and the co-occurrence relationships of mobile genetics elements (MGEs) with ßRGs intensified, indicating the enhancement of ßRGs' mobility in gut microbiome when the fish suffered from cefotaxime contamination. Furthermore, the path of partial least squares path modeling (PLS-PM) gave an integral assessment on the specific causality of cefotaxime treatment to ßRG profiles, showing that cefotaxime-mediated ßRGs variation was most ascribed to the alteration of MGEs under cefotaxime stress, followed by bacterial community, functioning both direct influence as ßRG-hosts and indirect effects via affecting MGEs. Finally, pathogenic bacteria Aeromonas was identified as the critical host for multiple ßRGs in fish guts, and its ß-lactam resistance increased over the duration time of cefotaxime exposure, suggesting the potential spreading risks for the antibiotic-resistant pathogens from environmental ecosystems to clinic. Overall, our finding emphasized cefotaxime contamination in aquatic surroundings could enhance the ß-lactam resistance and its transmission mobility in fish bodies.


Assuntos
Bactérias , Cefotaxima , Microbioma Gastrointestinal , Resistência beta-Lactâmica , Cefotaxima/toxicidade , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/genética , Peixe-Zebra/microbiologia , Poluentes Químicos da Água/toxicidade , Resistência beta-Lactâmica/efeitos dos fármacos , Resistência beta-Lactâmica/genética , Sequências Repetitivas Dispersas/genética , Bactérias/efeitos dos fármacos , Bactérias/genética , Animais , Aeromonas/efeitos dos fármacos , Aeromonas/genética
18.
Autophagy ; 19(1): 324-337, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35775203

RESUMO

The increasing prevalence of antifungal-resistant human pathogenic fungi, particularly azole-resistant Aspergillus fumigatus, is a life-threatening challenge to the immunocompromised population. Autophagy-related processes such as LC3-associated phagocytosis have been shown to be activated in the host response against fungal infection, but their overall effect on host resistance remains uncertain. To analyze the relevance of these processes in vivo, we used a zebrafish animal model of invasive Aspergillosis. To confirm the validity of this model to test potential treatments for this disease, we confirmed that immunosuppressive treatments or neutropenia rendered zebrafish embryos more susceptible to A. fumigatus. We used GFP-Lc3 transgenic zebrafish to visualize the autophagy-related processes in innate immune phagocytes shortly after phagocytosis of A. fumigatus conidia, and found that both wild-type and melanin-deficient conidia elicited Lc3 recruitment. In macrophages, we observed GFP-Lc3 accumulation in puncta after phagocytosis, as well as short, rapid events of GFP-Lc3 decoration of single and multiple conidia-containing vesicles, while neutrophils covered single conidia-containing vesicles with bright and long-lasting GFP-Lc3 signal. Next, using genetic and pharmacological stimulation of three independent autophagy-inducing pathways, we showed that the antifungal autophagy response improves the host survival against A. fumigatus infection, but only in the presence of phagocytes. Therefore, we provide proof-of-concept that stimulating the (auto)phagolysosomal pathways is a promising approach to develop host-directed therapies against invasive Aspergillosis, and should be explored further either as adjunctive or stand-alone therapy for drug-resistant Aspergillus infections.Abbreviations: DMSO: dimethyl sulfoxide; HR: hazard ratio; HDT: host-directed therapy; Hpf: hours post fertilization; IA: invasive Aspergillosis; LAP: LC3-associated phagocytosis; MTZ: metronidazole; PTU: N-phenylthiourea; ROS: reactive oxygen species.


Assuntos
Aspergilose , Peixe-Zebra , Animais , Humanos , Peixe-Zebra/microbiologia , Autofagia , Antifúngicos/farmacologia , Aspergilose/microbiologia , Fagocitose , Lisossomos , Esporos Fúngicos
19.
Neuropharmacology ; 225: 109401, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36565853

RESUMO

Certain bacteria possess the ability to reduce anxiety- and stress-related behaviors through the gut microbiome-brain axis. Such bacteria are called psychobiotics, and can be used to improve mood and cognition. However, only a few bacteria have been characterized as psychobiotics, and their exact mechanism of action remains unclear. Hence, in this study we analyzed three different species under the Lactobacillacea family, namely, Lactobacillus delbrueckii, Lacticaseibacillus casei, and Lacticaseibacillus paracasei for their potential psychobiotic activities. L. delbrueckii treatment reduced anxiety-like behavior and increased brain and gut glutamic acid decarboxylase (gad) gene expression in zebrafish. It also altered zebrafish gut microbial community as determined by PCR-DGGE and 16S rRNA-based metagenomics analysis. Overall, this paper showed that L. delbrueckii but not L. paracasei and L. casei, induced a consistent improvement in anxiety-like behavior in zebrafish, implicating its potential role as a psychobiotic to reduce anxiety. This article is part of the Special Issue on 'Microbiome & the Brain: Mechanisms & Maladies'.


Assuntos
Microbioma Gastrointestinal , Lactobacillus delbrueckii , Animais , Lactobacillus delbrueckii/genética , Lactobacillus delbrueckii/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/microbiologia , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/metabolismo , Encéfalo/metabolismo , Ansiedade
20.
Cell Rep ; 41(11): 111817, 2022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-36516756

RESUMO

During mycobacterial infections, pathogenic mycobacteria manipulate both host immune and stromal cells to establish and maintain a productive infection. In humans, non-human primates, and zebrafish models of infection, pathogenic mycobacteria produce and modify the specialized lipid trehalose 6,6'-dimycolate (TDM) in the bacterial cell envelope to drive host angiogenesis toward the site of forming granulomas, leading to enhanced bacterial growth. Here, we use the zebrafish-Mycobacterium marinum infection model to define the signaling basis of the host angiogenic response. Through intravital imaging and cell-restricted peptide-mediated inhibition, we identify macrophage-specific activation of NFAT signaling as essential to TDM-mediated angiogenesis in vivo. Exposure of cultured human cells to Mycobacterium tuberculosis results in robust induction of VEGFA, which is dependent on a signaling pathway downstream of host TDM detection and culminates in NFATC2 activation. As granuloma-associated angiogenesis is known to serve bacterial-beneficial roles, these findings identify potential host targets to improve tuberculosis disease outcomes.


Assuntos
Mycobacterium marinum , Mycobacterium tuberculosis , Tuberculose , Animais , Humanos , Peixe-Zebra/microbiologia , Macrófagos/metabolismo , Transdução de Sinais , Granuloma/patologia , Fatores de Transcrição NFATC/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...