RESUMO
The gut microbiota is a crucial regulator of anti-tumour immunity during immune checkpoint inhibitor therapy. Several bacteria that promote an anti-tumour response to immune checkpoint inhibitors have been identified in mice1-6. Moreover, transplantation of faecal specimens from responders can improve the efficacy of anti-PD-1 therapy in patients with melanoma7,8. However, the increased efficacy from faecal transplants is variable and how gut bacteria promote anti-tumour immunity remains unclear. Here we show that the gut microbiome downregulates PD-L2 expression and its binding partner repulsive guidance molecule b (RGMb) to promote anti-tumour immunity and identify bacterial species that mediate this effect. PD-L1 and PD-L2 share PD-1 as a binding partner, but PD-L2 can also bind RGMb. We demonstrate that blockade of PD-L2-RGMb interactions can overcome microbiome-dependent resistance to PD-1 pathway inhibitors. Antibody-mediated blockade of the PD-L2-RGMb pathway or conditional deletion of RGMb in T cells combined with an anti-PD-1 or anti-PD-L1 antibody promotes anti-tumour responses in multiple mouse tumour models that do not respond to anti-PD-1 or anti-PD-L1 alone (germ-free mice, antibiotic-treated mice and even mice colonized with stool samples from a patient who did not respond to treatment). These studies identify downregulation of the PD-L2-RGMb pathway as a specific mechanism by which the gut microbiota can promote responses to PD-1 checkpoint blockade. The results also define a potentially effective immunological strategy for treating patients who do not respond to PD-1 cancer immunotherapy.
Assuntos
Resistencia a Medicamentos Antineoplásicos , Imunoterapia , Melanoma , Microbiota , Animais , Humanos , Camundongos , Moléculas de Adesão Celular Neuronais , Modelos Animais de Doenças , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Transplante de Microbiota Fecal , Vida Livre de Germes , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Melanoma/imunologia , Melanoma/microbiologia , Melanoma/terapia , Ligação Proteica/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologiaRESUMO
Differentiating the effects of infant microbiota, developmental, and nutritional changes on immunological maturation during weaning is an ongoing challenge. In this issue of Cell Host & Microbe, Lubin and colleagues report a gnotobiotic mouse model that maintains neonatal-like microbiome composition into adulthood to help answer burning questions in this field.
Assuntos
Microbiota , Animais , Camundongos , Vida Livre de Germes , DesmameRESUMO
Disruptions to the intestinal microbiome during weaning lead to negative effects on host immune function. However, the critical host-microbe interactions during weaning that are required for immune system development remain poorly understood. We find that restricting microbiome maturation during weaning stunts immune system development and increases susceptibility to enteric infection. We developed a gnotobiotic mouse model of the early-life microbiome Pediatric Community (PedsCom). These mice develop fewer peripheral regulatory T cells and less IgA, hallmarks of microbiota-driven immune system development. Furthermore, adult PedsCom mice retain high susceptibility to Salmonella infection, which is characteristic of young mice and children. Altogether, our work illustrates how the post-weaning transition in microbiome composition contributes to normal immune maturation and protection from infection. Accurate modeling of the pre-weaning microbiome provides a window into the microbial requirements for healthy development and suggests an opportunity to design microbial interventions at weaning to improve immune development in human infants.
Assuntos
Microbioma Gastrointestinal , Microbiota , Lactente , Adulto , Animais , Humanos , Camundongos , Criança , Vida Livre de Germes , Desmame , Sistema ImunitárioRESUMO
Therapeutic manipulation of the gut microbiota holds great potential for human health. The mechanisms bacteria use to colonize the gut therefore present valuable targets for clinical intervention. We now report that bacteria use phase separation to enhance fitness in the mammalian gut. We establish that the intrinsically disordered region (IDR) of the broadly and highly conserved transcription termination factor Rho is necessary and sufficient for phase separation in vivo and in vitro in the human commensal Bacteroides thetaiotaomicron. Phase separation increases transcription termination by Rho in an IDR-dependent manner. Moreover, the IDR is critical for gene regulation in the gut. Our findings expose phase separation as vital for host-commensal bacteria interactions and relevant for novel clinical applications.
Assuntos
Proteínas de Bactérias , Bacteroides thetaiotaomicron , Microbioma Gastrointestinal , Aptidão Genética , Proteínas Intrinsicamente Desordenadas , RNA Helicases , Fator Rho , Animais , Humanos , Bacteroides thetaiotaomicron/genética , Bacteroides thetaiotaomicron/fisiologia , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiologia , Proteínas Intrinsicamente Desordenadas/química , Proteínas Intrinsicamente Desordenadas/genética , Proteínas Intrinsicamente Desordenadas/fisiologia , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , RNA Helicases/química , RNA Helicases/genética , RNA Helicases/fisiologia , Fator Rho/química , Fator Rho/genética , Fator Rho/fisiologia , Terminação da Transcrição Genética , Domínios Proteicos , Camundongos , Vida Livre de Germes , Camundongos Endogâmicos C57BL , Masculino , FemininoRESUMO
OBJECTIVE: Faecal microbiota transplantation (FMT) in germ-free (GF) mice is a common approach to study the causal role of the gut microbiota in metabolic diseases. Lack of consideration of housing conditions post-FMT may contribute to study heterogeneity. We compared the impact of two housing strategies on the metabolic outcomes of GF mice colonised by gut microbiota from mice treated with a known gut modulator (cranberry proanthocyanidins (PAC)) or vehicle. DESIGN: High-fat high-sucrose diet-fed GF mice underwent FMT-PAC colonisation in sterile individual positive flow ventilated cages under rigorous housing conditions and then maintained for 8 weeks either in the gnotobiotic-axenic sector or in the specific pathogen free (SPF) sector of the same animal facility. RESULTS: Unexpectedly, 8 weeks after colonisation, we observed opposing liver phenotypes dependent on the housing environment of mice. Mice housed in the GF sector receiving the PAC gut microbiota showed a significant decrease in liver weight and hepatic triglyceride accumulation compared with control group. Conversely, exacerbated liver steatosis was observed in the FMT-PAC mice housed in the SPF sector. These phenotypic differences were associated with housing-specific profiles of colonising bacterial in the gut and of faecal metabolites. CONCLUSION: These results suggest that the housing environment in which gnotobiotic mice are maintained post-FMT strongly influences gut microbiota composition and function and can lead to distinctive phenotypes in recipient mice. Better standardisation of FMT experiments is needed to ensure reproducible and translatable results.
Assuntos
Habitação , Microbiota , Animais , Camundongos , Qualidade Habitacional , Obesidade/metabolismo , Transplante de Microbiota Fecal , Fenótipo , Dieta Hiperlipídica/efeitos adversos , Vida Livre de Germes , Camundongos Endogâmicos C57BLRESUMO
Commensal bacteria are major contributors to mammalian metabolism. We used liquid chromatography mass spectrometry to study the metabolomes of germ-free, gnotobiotic, and specific-pathogen-free mice, while also evaluating the influence of age and sex on metabolite profiles. Microbiota modified the metabolome of all body sites and accounted for the highest proportion of variation within the gastrointestinal tract. Microbiota and age explained similar amounts of variation the metabolome of urine, serum, and peritoneal fluid, while age was the primary driver of variation in the liver and spleen. Although sex explained the least amount of variation at all sites, it had a significant impact on all sites except the ileum. Collectively, these data illustrate the interplay between microbiota, age, and sex in the metabolic phenotypes of diverse body sites. This provides a framework for interpreting complex metabolic phenotypes and will help guide future studies into the role that the microbiome plays in disease.
Assuntos
Metaboloma , Microbiota , Camundongos , Animais , Trato Gastrointestinal/microbiologia , Vida Livre de Germes , Organismos Livres de Patógenos Específicos , Metabolômica/métodos , MamíferosRESUMO
Pancreatic ductal adenocarcinoma (PDAC) is expected to be the second most deadly cancer by 2040, owing to the high incidence of metastatic disease and limited responses to treatment1,2. Less than half of all patients respond to the primary treatment for PDAC, chemotherapy3,4, and genetic alterations alone cannot explain this5. Diet is an environmental factor that can influence the response to therapies, but its role in PDAC is unclear. Here, using shotgun metagenomic sequencing and metabolomic screening, we show that the microbiota-derived tryptophan metabolite indole-3-acetic acid (3-IAA) is enriched in patients who respond to treatment. Faecal microbiota transplantation, short-term dietary manipulation of tryptophan and oral 3-IAA administration increase the efficacy of chemotherapy in humanized gnotobiotic mouse models of PDAC. Using a combination of loss- and gain-of-function experiments, we show that the efficacy of 3-IAA and chemotherapy is licensed by neutrophil-derived myeloperoxidase. Myeloperoxidase oxidizes 3-IAA, which in combination with chemotherapy induces a downregulation of the reactive oxygen species (ROS)-degrading enzymes glutathione peroxidase 3 and glutathione peroxidase 7. All of this results in the accumulation of ROS and the downregulation of autophagy in cancer cells, which compromises their metabolic fitness and, ultimately, their proliferation. In humans, we observed a significant correlation between the levels of 3-IAA and the efficacy of therapy in two independent PDAC cohorts. In summary, we identify a microbiota-derived metabolite that has clinical implications in the treatment of PDAC, and provide a motivation for considering nutritional interventions during the treatment of patients with cancer.
Assuntos
Carcinoma Ductal Pancreático , Microbiota , Neoplasias Pancreáticas , Animais , Humanos , Camundongos , Carcinoma Ductal Pancreático/dietoterapia , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/microbiologia , Glutationa Peroxidase/metabolismo , Neoplasias Pancreáticas/dietoterapia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/microbiologia , Peroxidase/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Triptofano/metabolismo , Triptofano/farmacologia , Triptofano/uso terapêutico , Neutrófilos/enzimologia , Autofagia , Metagenoma , Metabolômica , Transplante de Microbiota Fecal , Ácidos Indolacéticos/farmacologia , Ácidos Indolacéticos/uso terapêutico , Modelos Animais de Doenças , Vida Livre de GermesRESUMO
The intestinal microbiota is known to influence postnatal growth. We previously found that a strain of Lactiplantibacillus plantarum (strain LpWJL) buffers the adverse effects of chronic undernutrition on the growth of juvenile germ-free mice. Here, we report that LpWJL sustains the postnatal growth of malnourished conventional animals and supports both insulin-like growth factor-1 (IGF-1) and insulin production and activity. We have identified cell walls isolated from LpWJL, as well as muramyl dipeptide and mifamurtide, as sufficient cues to stimulate animal growth despite undernutrition. Further, we found that NOD2 is necessary in intestinal epithelial cells for LpWJL-mediated IGF-1 production and for postnatal growth promotion in malnourished conventional animals. These findings indicate that, coupled with renutrition, bacteria cell walls or purified NOD2 ligands have the potential to alleviate stunting.
Assuntos
Microbioma Gastrointestinal , Crescimento , Intestinos , Lactobacillaceae , Desnutrição , Proteína Adaptadora de Sinalização NOD2 , Animais , Camundongos , Parede Celular/química , Células Epiteliais/microbiologia , Células Epiteliais/fisiologia , Microbioma Gastrointestinal/fisiologia , Vida Livre de Germes , Transtornos do Crescimento/fisiopatologia , Transtornos do Crescimento/terapia , Insulina/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/fisiologia , Intestinos/microbiologia , Intestinos/fisiologia , Lactobacillaceae/fisiologia , Desnutrição/fisiopatologia , Desnutrição/terapia , Proteína Adaptadora de Sinalização NOD2/metabolismo , Crescimento/efeitos dos fármacos , Crescimento/fisiologia , Acetilmuramil-Alanil-Isoglutamina/farmacologia , Acetilmuramil-Alanil-Isoglutamina/uso terapêuticoRESUMO
Gnotobiotic (GN) animals with defined microbiota allow us to study host-microbiota and microbiota-microbiota interferences. Preterm germ-free (GF) piglets were mono-associated with probiotic Bifidobacterium animalis subsp. lactis BB-12 (BB12) to ameliorate/prevent the consequences of infection with the Salmonella Typhimurium strain LT2 (LT2). Goblet cell density; expression of Toll-like receptors (TLRs) 2, 4, and 9; high mobility group box 1 (HMGB1); interleukin (IL)-6; and IL-12/23p40 were analyzed to evaluate the possible modulatory effect of BB12. BB12 prevented an LT2-induced decrease of goblet cell density in the colon. TLRs signaling modified by LT2 was not influenced by the previous association with BB12. The expression of HMGB1, IL-6, and IL12/23p40 in the jejunum, ileum, and colon and their levels in plasma were all decreased by BB12, but these changes were not statistically significant. In the colon, differences in HMGB1 distribution between the GF and LT2 piglet groups were observed. In conclusion, the mono-association of GF piglets with BB12 prior to LT2 infection partially ameliorated the inflammatory response to LT2 infection.
Assuntos
Bifidobacterium animalis , Proteína HMGB1 , Probióticos , Animais , Humanos , Recém-Nascido , Vida Livre de Germes , Recém-Nascido Prematuro , Probióticos/farmacologia , Salmonella typhimurium , Suínos , Receptores Toll-Like/metabolismoRESUMO
Human rotavirus (HRV) is a leading cause of gastroenteritis in children under 5 years of age. Licensed vaccines containing G1P[8] and G1-4P[8] strains are less efficacious against newly emerging P[6] strains, indicating an urgent need for better cross protective vaccines. Here, we report our development of a new gnotobiotic (Gn) pig model of P[6] HRV infection and disease as a tool for evaluating potential vaccine candidates. The Arg HRV (G4P[6]) strain was derived from a diarrheic human infant stool sample and determined to be free of other viruses by metagenomic sequencing. Neonatal Gn pigs were orally inoculated with the stool suspension containing 5.6 × 105 fluorescent focus units (FFU) of the virus. Small and large intestinal contents were collected at post inoculation day 2 or 3. The virus was passaged 6 times in neonatal Gn pigs to generate a large inoculum pool. Next, 33-34 day old Gn pigs were orally inoculated with 10-2, 103, 104, and 105 FFU of Arg HRV to determine the optimal challenge dose. All pigs developed clinical signs of infection, regardless of the inoculum dose. The optimal challenge dose was determined to be 105 FFU. This new Gn pig model is ready to be used to assess the protective efficacy of candidate monovalent and multivalent vaccines against P[6] HRV.
Assuntos
Infecções por Rotavirus , Vacinas contra Rotavirus , Rotavirus , Lactente , Criança , Recém-Nascido , Humanos , Animais , Suínos , Pré-Escolar , Infecções por Rotavirus/prevenção & controle , Infecções por Rotavirus/veterinária , Fezes , Vida Livre de GermesRESUMO
Human rotavirus (HRV) is a major cause of childhood diarrhea in developing countries where widespread malnutrition contributes to the decreased oral vaccine efficacy and increased prevalence of other enteric infections, which are major concerns for global health. Neonatal gnotobiotic (Gn) piglets closely resemble human infants in their anatomy, physiology, and outbred status, providing a unique model to investigate malnutrition, supplementations, and HRV infection. To understand the molecular signatures associated with immune enhancement and reduced diarrheal severity by Escherichia coli Nissle 1917 (EcN) and tryptophan (TRP), immunological responses and global nontargeted metabolomics and lipidomics approaches were investigated on the plasma and fecal contents of malnourished pigs transplanted with human infant fecal microbiota and infected with virulent (Vir) HRV. Overall, EcN + TRP combined (rather than individual supplement action) promoted greater and balanced immunoregulatory/immunostimulatory responses associated with greater protection against HRV infection and disease in malnourished humanized piglets. Moreover, EcN + TRP treatment upregulated the production of several metabolites with immunoregulatory/immunostimulatory properties: amino acids (N-acetylserotonin, methylacetoacetyl-CoA), lipids (gamma-butyrobetaine, eicosanoids, cholesterol-sulfate, sphinganine/phytosphingosine, leukotriene), organic compound (biliverdin), benzenoids (gentisic acid, aminobenzoic acid), and nucleotides (hypoxathine/inosine/xanthine, cytidine-5'-monophosphate). Additionally, the levels of several proinflammatory metabolites of organic compounds (adenosylhomocysteine, phenylacetylglycine, urobilinogen/coproporphyrinogen) and amino acid (phenylalanine) were reduced following EcN + TRP treatment. These results suggest that the EcN + TRP effects on reducing HRV diarrhea in neonatal Gn pigs were at least in part due to altered metabolites, those involved in lipid, amino acid, benzenoids, organic compounds, and nucleotide metabolism. Identification of these important mechanisms of EcN/TRP prevention of HRV diarrhea provides novel targets for therapeutics development. IMPORTANCE Human rotavirus (HRV) is the most common cause of viral gastroenteritis in children, especially in developing countries, where the efficacy of oral HRV vaccines is reduced. Escherichia coli Nissle 1917 (EcN) is used to treat enteric infections and ulcerative colitis while tryptophan (TRP) is a biomarker of malnutrition, and its supplementation can alleviate intestinal inflammation and normalize intestinal microbiota in malnourished hosts. Supplementation of EcN + TRP to malnourished humanized gnotobiotic piglets enhanced immune responses and resulted in greater protection against HRV infection and diarrhea. Moreover, EcN + TRP supplementation increased the levels of immunoregulatory/immunostimulatory metabolites while decreasing the production of proinflammatory metabolites in plasma and fecal samples. Profiling of immunoregulatory and proinflammatory biomarkers associated with HRV perturbations will aid in the identification of treatments against HRV and other enteric diseases in malnourished children.
Assuntos
Infecções por Escherichia coli , Transplante de Microbiota Fecal , Desnutrição , Infecções por Rotavirus , Triptofano , Animais , Humanos , Lactente , Aminobenzoatos , Biliverdina/metabolismo , Colesterol , Coenzima A/metabolismo , Coproporfirinogênios , Citidina/metabolismo , Diarreia , Escherichia coli/metabolismo , Vida Livre de Germes , Inosina/metabolismo , Lipídeos , Desnutrição/terapia , Desnutrição/complicações , Metaboloma , Microbiota , Nucleotídeos/metabolismo , Fenilalanina/metabolismo , Rotavirus , Sulfatos , Suínos , Triptofano/farmacologia , Urobilinogênio/metabolismo , XantinasRESUMO
Interaction between the host and the enteric microbiome is highly complex. Microbial involvement in certain pathologies is moderately well established, but the contribution of the microbiome to animal welfare, behavior, sustainability, immune development, nutritional status, physiology, and maturation is less clear. A valuable experimental model to enable scientists to explore the role of the microbiome in various domains is to compare various phenotypes of a conventionally reared (CV) cohort with those in a germ-free (GF) state. A GF animal is one that is devoid of any detectable microbial life including bacteria, viruses, protozoa and parasites. The GF state is different from gnotobiotic animals where the microbiome is fully described, or 'specific pathogen free' (SPF) animals where a moderately normal microbiome is present but devoid of pathogenic microorganisms. Pioneering GF research in poultry in the late 1940s and 1950s has its origin in a need understand the mode of action of antibiotics. Early researchers quickly established that GF chicks responded differently to antibiotics than CV counterparts. The GF experimental model has since been exploited in many divergent fields including pathology, immunology, metabolism, anatomy, physiology, and others. The absence of a microbiome presents the host with a range of advantages and disadvantages. For example, GF chicks often grow more quickly and have lower feed conversion ratio (FCR) than their CV counterparts but may be less resilient to external stress and have a compromised immunological maturation rate. This review will summarize the literature on GF animal research with a special emphasis on poultry. The objective of the review is to establish a frame of reference to understand the extent of the role of the microbiome in animal health, welfare, nutrition, and growth, to provide opportunities for targeted modulation of the microbiome to achieve desired phenotypic responses whilst simultaneously minimizing unintended collateral effects.
Assuntos
Microbioma Gastrointestinal , Animais , Antibacterianos , Biologia , Galinhas , Microbioma Gastrointestinal/genética , Vida Livre de Germes , Humanos , Aves DomésticasRESUMO
Efforts to model the human gut microbiome in mice have led to important insights into the mechanisms of host-microbe interactions. However, the model communities studied to date have been defined or complex, but not both, limiting their utility. Here, we construct and characterize in vitro a defined community of 104 bacterial species composed of the most common taxa from the human gut microbiota (hCom1). We then used an iterative experimental process to fill open niches: germ-free mice were colonized with hCom1 and then challenged with a human fecal sample. We identified new species that engrafted following fecal challenge and added them to hCom1, yielding hCom2. In gnotobiotic mice, hCom2 exhibited increased stability to fecal challenge and robust colonization resistance against pathogenic Escherichia coli. Mice colonized by either hCom2 or a human fecal community are phenotypically similar, suggesting that this consortium will enable a mechanistic interrogation of species and genes on microbiome-associated phenotypes.
Assuntos
Microbioma Gastrointestinal , Microbiota , Animais , Bactérias/genética , Escherichia coli , Fezes , Microbioma Gastrointestinal/genética , Vida Livre de Germes , Humanos , CamundongosRESUMO
Equipped with a novel isolator-housed metabolic cage system, a study in PLOS Biology assessed how the metabolism of mice harboring a defined minimal microbial community (OligoMM12) differs from that of germ-free and conventionally colonized mice.
Assuntos
Microbioma Gastrointestinal , Microbiota , Animais , Vida Livre de Germes , CamundongosRESUMO
The capacity of the intestinal microbiota to degrade otherwise indigestible diet components is known to greatly improve the recovery of energy from food. This has led to the hypothesis that increased digestive efficiency may underlie the contribution of the microbiota to obesity. OligoMM12-colonized gnotobiotic mice have a consistently higher fat mass than germ-free (GF) or fully colonized counterparts. We therefore investigated their food intake, digestion efficiency, energy expenditure, and respiratory quotient using a novel isolator-housed metabolic cage system, which allows long-term measurements without contamination risk. This demonstrated that microbiota-released calories are perfectly balanced by decreased food intake in fully colonized versus gnotobiotic OligoMM12 and GF mice fed a standard chow diet, i.e., microbiota-released calories can in fact be well integrated into appetite control. We also observed no significant difference in energy expenditure after normalization by lean mass between the different microbiota groups, suggesting that cumulative small differences in energy balance, or altered energy storage, must underlie fat accumulation in OligoMM12 mice. Consistent with altered energy storage, major differences were observed in the type of respiratory substrates used in metabolism over the circadian cycle: In GF mice, the respiratory exchange ratio (RER) was consistently lower than that of fully colonized mice at all times of day, indicative of more reliance on fat and less on glucose metabolism. Intriguingly, the RER of OligoMM12-colonized gnotobiotic mice phenocopied fully colonized mice during the dark (active/eating) phase but phenocopied GF mice during the light (fasting/resting) phase. Further, OligoMM12-colonized mice showed a GF-like drop in liver glycogen storage during the light phase and both liver and plasma metabolomes of OligoMM12 mice clustered closely with GF mice. This implies the existence of microbiota functions that are required to maintain normal host metabolism during the resting/fasting phase of circadian cycle and which are absent in the OligoMM12 consortium.
Assuntos
Glicogênio Hepático , Microbiota , Animais , Vida Livre de Germes , Glucose , Camundongos , Obesidade/metabolismoRESUMO
Indole signaling is viewed as a potential target for antivirulence therapy against antibiotic-resistant pathogens because of its link with the production of virulence factors. This study examined the antimicrobial and antivirulence properties of 44 indoles toward Vibrio campbellii. Based on the results, 17 halogenated indole analogues were selected, as they significantly improved the survival of brine shrimp larvae challenged with V. campbellii. Specifically, 6-bromoindole, 7-bromoindole, 4-fluoroindole, 5-iodoindole, and 7-iodoindole showed a high protective effect, improving the survival of brine shrimp to over 80% even at a low concentration of 10 µM. To explore the impact of selected indole analogues on bacterial virulence phenotypes, swimming motility, biofilm formation, protease activity, and hemolytic activity of V. campbellii were determined. The results showed that all of the 17 selected indole analogues decreased swimming motility at both 10 µM and 100 µM. Most of the indole analogues decreased biofilm formation at a concentration of 100 µM. In contrast, only a slightly decreased protease activity and no effect on hemolytic activity were observed at both concentrations. To our knowledge, this is the first study of the structure-activity relation of halogenated indole analogues with respect to virulence inhibition of a pathogenic bacterium in an in vivo host model system, and the results demonstrate the potential of these compounds in applications aiming at the protection of shrimp from vibriosis, a major disease in aquaculture. IMPORTANCE Bacterial diseases are a major problem in the aquaculture industry. In order to counter this problem, farmers have been using antibiotics, and this has led to the evolution and spread of antibiotic resistance. In order for the aquaculture industry to further grow in a sustainable way, novel and sustainable methods to control diseases are needed. We previously reported that indole signaling is a valid target for the development of novel therapies to control disease caused by Vibrio campbellii and related bacteria, which are among the major bacterial pathogens in aquaculture. In the present study, we identified indole analogues that are more potent in protecting brine shrimp (a model organism for shrimp) from V. campbellii. To our knowledge, this is the first study of the structure-activity relation of halogenated indole analogues with respect to virulence inhibition of a pathogenic bacterium in an in vivo host model system.
Assuntos
Anti-Infecciosos , Vibrio , Animais , Antibacterianos/farmacologia , Anti-Infecciosos/farmacologia , Artemia/microbiologia , Vida Livre de Germes , Indóis/farmacologia , Peptídeo Hidrolases/farmacologia , Virulência/genética , Fatores de Virulência/genéticaRESUMO
SCOPE: The gut microbiota plays a prominent role in gut-brain interactions and gut dysbiosis is involved in neuroinflammation. However, specific probiotics targeting neuroinflammation need to be explored. In this study, the antineuroinflammatory effect of the potential probiotic Roseburia hominis (R. hominis) and its underlying mechanisms is investigated. METHODS AND RESULTS: First, germ-free (GF) rats are orally treated with R. hominis. Microglial activation, proinflammatory cytokines, levels of short-chain fatty acids, depressive behaviors, and visceral sensitivity are assessed. Second, GF rats are treated with propionate or butyrate, and microglial activation, proinflammatory cytokines, histone deacetylase 1 (HDAC1), and histone H3 acetyl K9 (Ac-H3K9) are analyzed. The results show that R. hominis administration inhibits microglial activation, reduces the levels of IL-1α, INF-γ, and MCP-1 in the brain, and alleviates depressive behaviors and visceral hypersensitivity in GF rats. Moreover, the serum levels of propionate and butyrate are increased significantly in the R. hominis-treated group. Propionate or butyrate treatment reduces microglial activation, the levels of proinflammatory cytokines and HDAC1, and promotes the expression of Ac-H3K9 in the brain. CONCLUSION: These findings suggest that R. hominis alleviates neuroinflammation by producing propionate and butyrate, which serve as HDAC inhibitors. This study provides a potential psychoprobiotic to reduce neuroinflammation.
Assuntos
Eixo Encéfalo-Intestino , Butiratos , Clostridiales , Ácidos Graxos Voláteis , Histona Desacetilase 1 , Doenças Neuroinflamatórias , Probióticos , Propionatos , Animais , Butiratos/sangue , Butiratos/metabolismo , Clostridiales/metabolismo , Citocinas/metabolismo , Ácidos Graxos Voláteis/metabolismo , Vida Livre de Germes , Histona Desacetilase 1/metabolismo , Histonas/metabolismo , Doenças Neuroinflamatórias/terapia , Probióticos/uso terapêutico , Propionatos/sangue , Propionatos/metabolismo , RatosRESUMO
The cost of supply cylinders can be expensive for smaller gnotobiotic facilities on strict budgets. Here, we present an alternative approach to enter supplies into germ-free isolators that does not require large cylinder drums (supply cylinders). We describe procedures for autoclaving double-bagged packs of supplies, sealing the packs aseptically into zip lock bags in biosafety cabinet, and entering supply packages into isolators. Our gnotobiotic facility has been using this protocol since 2018 and has been effective in maintaining germ-free status.
Assuntos
Vida Livre de Germes , Roedores , Animais , Bandagens , Embalagem de Medicamentos , QuarentenaRESUMO
Plant fibers in byproduct streams produced by non-harsh food processing methods represent biorepositories of diverse, naturally occurring, and physiologically active biomolecules. To demonstrate one approach for their characterization, mass spectrometry of intestinal contents from gnotobiotic mice, plus in vitro studies, revealed liberation of N-methylserotonin from orange fibers by human gut microbiota members including Bacteroides ovatus. Functional genomic analyses of B. ovatus strains grown under permissive and non-permissive N-methylserotonin "mining" conditions revealed polysaccharide utilization loci that target pectins whose expression correlate with strain-specific liberation of this compound. N-methylserotonin, orally administered to germ-free mice, reduced adiposity, altered liver glycogenesis, shortened gut transit time, and changed expression of genes that regulate circadian rhythm in the liver and colon. In human studies, dose-dependent, orange-fiber-specific fecal accumulation of N-methylserotonin positively correlated with levels of microbiome genes encoding enzymes that digest pectic glycans. Identifying this type of microbial mining activity has potential therapeutic implications.
Assuntos
Citrus sinensis , Microbioma Gastrointestinal , Animais , Citrus sinensis/metabolismo , Fibras na Dieta , Microbioma Gastrointestinal/fisiologia , Vida Livre de Germes , Humanos , Camundongos , Pectinas/metabolismo , Polissacarídeos/metabolismo , Serotonina/análogos & derivadosRESUMO
Gut bacteria influence the brain and behavior, but causation in humans remains unclear.