Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 15: 1347676, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38590519

RESUMO

The gut-lung axis is critical during viral respiratory infections such as influenza. Gut dysbiosis during infection translates into a massive drop of microbially produced short-chain fatty acids (SCFAs). Among them, butyrate is important during influenza suggesting that microbiome-based therapeutics targeting butyrate might hold promises. The butyrate-producing bacterium Faecalibacterium duncaniae (formerly referred to as F. prausnitzii) is an emerging probiotic with several health-promoting characteristics. To investigate the potential effects of F. duncaniae on influenza outcomes, mice were gavaged with live F. duncaniae (A2-165 or I-4574 strains) five days before infection. Supplementation of F. duncaniae was associated with less severe disease, a lower pulmonary viral load, and lower levels of lung inflammation. F. duncaniae supplementation impacted on gut dysbiosis induced by infection, as assessed by 16S rRNA sequencing. Interestingly, F. duncaniae administration was associated with a recovery in levels of SCFAs (including butyrate) in infected animals. The live form of F. duncaniae was more potent that the pasteurized form in improving influenza outcomes. Lastly, F. duncaniae partially protected against secondary (systemic) bacterial infection. We conclude that F. duncaniae might serve as a novel next generation probiotic against acute viral respiratory diseases.


Assuntos
Influenza Humana , Probióticos , Camundongos , Animais , Humanos , Disbiose/microbiologia , RNA Ribossômico 16S/genética , Ácidos Graxos Voláteis , Butiratos , Faecalibacterium/genética
2.
Gut Microbes ; 16(1): 2325067, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38445660

RESUMO

The gut-to-lung axis is critical during respiratory infections, including influenza A virus (IAV) infection. In the present study, we used high-resolution shotgun metagenomics and targeted metabolomic analysis to characterize influenza-associated changes in the composition and metabolism of the mouse gut microbiota. We observed several taxonomic-level changes on day (D)7 post-infection, including a marked reduction in the abundance of members of the Lactobacillaceae and Bifidobacteriaceae families, and an increase in the abundance of Akkermansia muciniphila. On D14, perturbation persisted in some species. Functional scale analysis of metagenomic data revealed transient changes in several metabolic pathways, particularly those leading to the production of short-chain fatty acids (SCFAs), polyamines, and tryptophan metabolites. Quantitative targeted metabolomics analysis of the serum revealed changes in specific classes of gut microbiota metabolites, including SCFAs, trimethylamine, polyamines, and indole-containing tryptophan metabolites. A marked decrease in indole-3-propionic acid (IPA) blood level was observed on D7. Changes in microbiota-associated metabolites correlated with changes in taxon abundance and disease marker levels. In particular, IPA was positively correlated with some Lactobacillaceae and Bifidobacteriaceae species (Limosilactobacillus reuteri, Lactobacillus animalis) and negatively correlated with Bacteroidales bacterium M7, viral load, and inflammation markers. IPA supplementation in diseased animals reduced viral load and lowered local (lung) and systemic inflammation. Treatment of mice with antibiotics targeting IPA-producing bacteria before infection enhanced viral load and lung inflammation, an effect inhibited by IPA supplementation. The results of this integrated metagenomic-metabolomic analysis highlighted IPA as an important contributor to influenza outcomes and a potential biomarker of disease severity.


Assuntos
Actinobacteria , Microbioma Gastrointestinal , Influenza Humana , Humanos , Animais , Camundongos , Propionatos , Triptofano , Inflamação , Poliaminas
3.
Cells ; 12(9)2023 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-37174660

RESUMO

The gut microbiota is now considered as a key player in the development of metabolic dysfunction. Therefore, targeting gut microbiota dysbiosis has emerged as a new therapeutic strategy, notably through the use of live gut microbiota-derived biotherapeutics. We previously highlighted the anti-inflammatory abilities of two Parabacteroides distasonis strains. We herein evaluate their potential anti-obesity abilities and show that the two strains induced the secretion of the incretin glucagon-like peptide 1 in vitro and limited weight gain and adiposity in obese mice. These beneficial effects are associated with reduced inflammation in adipose tissue and the improvement of lipid and bile acid metabolism markers. P. distasonis supplementation also modified the Actinomycetota, Bacillota and Bacteroidota taxa of the mice gut microbiota. These results provide better insight into the capacity of P. distasonis to positively influence host metabolism and to be used as novel source of live biotherapeutics in the treatment and prevention of metabolic-related diseases.


Assuntos
Microbioma Gastrointestinal , Obesidade , Animais , Camundongos , Obesidade/terapia , Obesidade/metabolismo , Bacteroidetes , Tecido Adiposo/metabolismo
4.
Front Immunol ; 13: 1024998, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36569929

RESUMO

Many parasitic diseases (including cerebral malaria, human African trypanosomiasis, cerebral toxoplasmosis, neurocysticercosis and neuroschistosomiasis) feature acute or chronic brain inflammation processes, which are often associated with deregulation of glial cell activity and disruption of the brain blood barrier's intactness. The inflammatory responses of astrocytes and microglia during parasite infection are strongly influenced by a variety of environmental factors. Although it has recently been shown that the gut microbiota influences the physiology and immunomodulation of the central nervous system in neurodegenerative diseases like Alzheimer's disease and Parkinson's, the putative link in parasite-induced neuroinflammatory diseases has not been well characterized. Likewise, the central nervous system can influence the gut microbiota. In parasite infections, the gut microbiota is strongly perturbed and might influence the severity of the central nervous system inflammation response through changes in the production of bacterial metabolites. Here, we review the roles of astrocytes and microglial cells in the neuropathophysiological processes induced by parasite infections and their possible regulation by the gut microbiota.


Assuntos
Microbioma Gastrointestinal , Malária Cerebral , Humanos , Microbioma Gastrointestinal/fisiologia , Doenças Neuroinflamatórias , Sistema Nervoso Central/metabolismo , Inflamação/metabolismo
5.
Sci Rep ; 12(1): 17591, 2022 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-36266398

RESUMO

Live biotherapeutic products constitute an emerging therapeutic approach to prevent or treat inflammatory bowel diseases. Lactobacillus acidophilus is a constituent of the human microbiota with probiotic potential, that is illustrated by improvement of intestinal inflammation and antimicrobial activity against several pathogens. In this study, we evaluated the immunomodulatory properties of the L. acidophilus strain BIO5768 at steady state and upon acute inflammation. Supplementation of naïve mice with BIO5768 heightened the transcript level of some IL-17 target genes encoding for protein with microbicidal activity independently of NOD2 signaling. Of these, the BIO5768-induced expression of Angiogenin-4 was blunted in monocolonized mice that are deficient for the receptor of IL-17 (but not for NOD2). Interestingly, priming of bone marrow derived dendritic cells by BIO5768 enhanced their ability to support the secretion of IL-17 by CD4+ T cells. Equally of importance, the production of IL-22 by type 3 innate lymphoid cells is concomitantly heightened in response to BIO5768. When administered alone or in combination with Bifidobacterium animalis spp. lactis BIO5764 and Limosilactobacillus reuteri, BIO5768 was able to alleviate at least partially intestinal inflammation induced by Citrobacter rodentium infection. Furthermore, BIO5768 was also able to improve colitis induced by 2,4,6-trinitrobenzene sulfonic acid (TNBS). In conclusion, we identify a new potential probiotic strain for the management of inflammatory bowel diseases, and provide some insights into its IL-17-dependent and independent mode of action.


Assuntos
Colite , Imunidade Inata , Doenças Inflamatórias Intestinais , Lactobacillus acidophilus , Probióticos , Animais , Camundongos , Bifidobacterium animalis , Colite/induzido quimicamente , Colite/terapia , Colite/microbiologia , Infecções por Enterobacteriaceae/terapia , Inflamação , Doenças Inflamatórias Intestinais/terapia , Interleucina-17 , Linfócitos , Probióticos/farmacologia , Probióticos/uso terapêutico , Ácido Trinitrobenzenossulfônico/efeitos adversos
6.
Nat Commun ; 13(1): 5324, 2022 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-36088459

RESUMO

Tissue injury triggers activation of mesenchymal lineage cells into wound-repairing myofibroblasts, whose unrestrained activity leads to fibrosis. Although this process is largely controlled at the transcriptional level, whether the main transcription factors involved have all been identified has remained elusive. Here, we report multi-omics analyses unraveling Basonuclin 2 (BNC2) as a myofibroblast identity transcription factor. Using liver fibrosis as a model for in-depth investigations, we first show that BNC2 expression is induced in both mouse and human fibrotic livers from different etiologies and decreases upon human liver fibrosis regression. Importantly, we found that BNC2 transcriptional induction is a specific feature of myofibroblastic activation in fibrotic tissues. Mechanistically, BNC2 expression and activities allow to integrate pro-fibrotic stimuli, including TGFß and Hippo/YAP1 signaling, towards induction of matrisome genes such as those encoding type I collagen. As a consequence, Bnc2 deficiency blunts collagen deposition in livers of mice fed a fibrogenic diet. Additionally, our work establishes BNC2 as potentially druggable since we identified the thalidomide derivative CC-885 as a BNC2 inhibitor. Altogether, we propose that BNC2 is a transcription factor involved in canonical pathways driving myofibroblastic activation in fibrosis.


Assuntos
Cirrose Hepática , Miofibroblastos , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Genômica , Humanos , Cirrose Hepática/genética , Cirrose Hepática/metabolismo , Camundongos , Miofibroblastos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
7.
Gut Microbes ; 14(1): 2100200, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35830432

RESUMO

Obese patientss with nonalcoholic steatohepatitis (NASH) are particularly prone to developing severe forms of coronavirus disease 19 (COVID-19). The gut-to-lung axis is critical during viral infections of the respiratory tract, and a change in the gut microbiota's composition might have a critical role in disease severity. Here, we investigated the consequences of infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on the gut microbiota in the context of obesity and NASH. To this end, we set up a nutritional model of obesity with dyslipidemia and NASH in the golden hamster, a relevant preclinical model of COVID-19. Relative to lean non-NASH controls, obese NASH hamsters develop severe inflammation of the lungs and liver. 16S rRNA gene profiling showed that depending on the diet, SARS-CoV-2 infection induced various changes in the gut microbiota's composition. Changes were more prominent and transient at day 4 post-infection in lean animals, alterations still persisted at day 10 in obese NASH animals. A targeted, quantitative metabolomic analysis revealed changes in the gut microbiota's metabolic output, some of which were diet-specific and regulated over time. Our results showed that specifically diet-associated taxa are correlated with disease parameters. Correlations between infection variables and diet-associated taxa highlighted a number of potentially protective or harmful bacteria in SARS-CoV-2-infected hamsters. In particular, some taxa in obese NASH hamsters (e.g. Blautia and Peptococcus) were associated with pro-inflammatory parameters in both the lungs and the liver. These taxon profiles and their association with specific disease markers suggest that microbial patterns might influence COVID-19 outcomes.


Assuntos
COVID-19 , Microbioma Gastrointestinal , Hepatopatia Gordurosa não Alcoólica , Animais , Cricetinae , Hepatopatia Gordurosa não Alcoólica/microbiologia , Obesidade/complicações , Obesidade/microbiologia , RNA Ribossômico 16S/genética , SARS-CoV-2
8.
Gut Microbes ; 14(1): 2018900, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34965194

RESUMO

Mounting evidence suggests that the gut-to-lung axis is critical during respiratory viral infections. We herein hypothesized that disruption of gut homeostasis during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection may associate with early disease outcomes. To address this question, we took advantage of the Syrian hamster model. Our data confirmed that this model recapitulates some hallmark features of the human disease in the lungs. We further showed that SARS-CoV-2 infection associated with mild intestinal inflammation, relative alteration in intestinal barrier property and liver inflammation and altered lipid metabolism. These changes occurred concomitantly with an alteration of the gut microbiota composition over the course of infection, notably characterized by a higher relative abundance of deleterious bacterial taxa such as Enterobacteriaceae and Desulfovibrionaceae. Conversely, several members of the Ruminococcaceae and Lachnospiraceae families, including bacteria known to produce the fermentative products short-chain fatty acids (SCFAs), had a reduced relative proportion compared to non-infected controls. Accordingly, infection led to a transient decrease in systemic SCFA amounts. SCFA supplementation during infection had no effect on clinical and inflammatory parameters. Lastly, a strong correlation between some gut microbiota taxa and clinical and inflammation indices of SARS-CoV-2 infection severity was evidenced. Collectively, alteration of the gut microbiota correlates with disease severity in hamsters making this experimental model valuable for the design of interventional, gut microbiota-targeted, approaches for the control of COVID-19.Abbreviations: SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; COVID-19, coronavirus disease 2019; SCFAs, short-chain fatty acids; dpi, day post-infection; RT-PCR, reverse transcription polymerase chain reaction; IL, interleukin. ACE2, angiotensin converting enzyme 2; TMPRSS2, transmembrane serine protease 2.


Assuntos
COVID-19/microbiologia , COVID-19/fisiopatologia , Modelos Animais de Doenças , Microbioma Gastrointestinal , Mesocricetus , Animais , Bactérias/classificação , Bactérias/isolamento & purificação , Bactérias/metabolismo , COVID-19/patologia , Cricetinae , Ácidos Graxos Voláteis/administração & dosagem , Ácidos Graxos Voláteis/metabolismo , Humanos , Masculino , SARS-CoV-2/fisiologia , Índice de Gravidade de Doença , Tratamento Farmacológico da COVID-19
10.
Microorganisms ; 9(3)2021 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-33803291

RESUMO

The role of the gut microbiota in health and disease is well recognized and the microbiota dysbiosis observed in many chronic diseases became a new therapeutic target. The challenge is to get a better insight into the functionality of commensal bacteria and to use this knowledge to select live biotherapeutics as new preventive or therapeutic products. In this study, we set up a screening approach to evaluate the functional capacities of a set of 21 strains isolated from the gut microbiota of neonates and adults. For this purpose, we selected key biological processes involved in the microbiome-host symbiosis and known to impact the host physiology i.e., the production of short-chain fatty acids and the ability to strengthen an epithelial barrier (Caco-2), to induce the release of the anti-inflammatory IL-10 cytokine after co-culture with human immune cells (PBMC) or to increase GLP-1 production from STC-1 endocrine cell line. This strategy highlighted fifteen strains exhibiting beneficial activities among which seven strains combined several of them. Interestingly, this work revealed for the first time a high prevalence of potential health-promoting functions among intestinal commensal strains and identified several appealing novel candidates for the management of chronic diseases, notably obesity and inflammatory bowel diseases.

11.
Infect Immun ; 89(9): e0073420, 2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-33820816

RESUMO

Along with respiratory tract disease per se, viral respiratory infections can also cause extrapulmonary complications with a potentially critical impact on health. In the present study, we used an experimental model of influenza A virus (IAV) infection to investigate the nature and outcome of the associated gut disorders. In IAV-infected mice, the signs of intestinal injury and inflammation, altered gene expression, and compromised intestinal barrier functions peaked on day 7 postinfection. As a likely result of bacterial component translocation, gene expression of inflammatory markers was upregulated in the liver. These changes occurred concomitantly with an alteration of the composition of the gut microbiota and with a decreased production of the fermentative, gut microbiota-derived products short-chain fatty acids (SCFAs). Gut inflammation and barrier dysfunction during influenza were not attributed to reduced food consumption, which caused in part gut dysbiosis. Treatment of IAV-infected mice with SCFAs was associated with an enhancement of intestinal barrier properties, as assessed by a reduction in the translocation of dextran and a decrease in inflammatory gene expression in the liver. Lastly, SCFA supplementation during influenza tended to reduce the translocation of the enteric pathogen Salmonella enterica serovar Typhimurium and to enhance the survival of doubly infected animals. Collectively, influenza virus infection can remotely impair the gut's barrier properties and trigger secondary enteric infections. The latter phenomenon can be partially countered by SCFA supplementation.


Assuntos
Infecções por Enterobacteriaceae/etiologia , Ácidos Graxos Voláteis/biossíntese , Interações Hospedeiro-Patógeno , Vírus da Influenza A/fisiologia , Influenza Humana/complicações , Influenza Humana/virologia , Mucosa Intestinal/metabolismo , Interações Microbianas , Suscetibilidade a Doenças , Disbiose , Infecções por Enterobacteriaceae/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Influenza Humana/metabolismo , Mucosa Intestinal/imunologia
12.
Nutrients ; 13(3)2021 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-33668212

RESUMO

Since alterations of the gut microbiota have been shown to play a major role in obesity, probiotics have attracted attention. Our aim was to identify probiotic candidates for the management of obesity using a combination of in vitro and in vivo approaches. We evaluated in vitro the ability of 23 strains to limit lipid accumulation in adipocytes and to enhance the secretion of satiety-promoting gut peptide in enteroendocrine cells. Following the in vitro screening, selected strains were further investigated in vivo, single, or as mixtures, using a murine model of diet-induced obesity. Strain Bifidobacterium longum PI10 administrated alone and the mixture of B. animalis subsp. lactis LA804 and Lactobacillus gasseri LA806 limited body weight gain and reduced obesity-associated metabolic dysfunction and inflammation. These protective effects were associated with changes in the hypothalamic gene expression of leptin and leptin receptor as well as with changes in the composition of gut microbiota and the profile of bile acids. This study provides crucial clues to identify new potential probiotics as effective therapeutic approaches in the management of obesity, while also providing some insights into their mechanisms of action.


Assuntos
Adipócitos/microbiologia , Células Enteroendócrinas/microbiologia , Microbioma Gastrointestinal/fisiologia , Obesidade/microbiologia , Probióticos/farmacologia , Animais , Ácidos e Sais Biliares/metabolismo , Dieta/efeitos adversos , Modelos Animais de Doenças , Hormônios Gastrointestinais/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Camundongos , Obesidade/etiologia , Manejo da Obesidade/métodos , Receptores para Leptina/metabolismo , Aumento de Peso/fisiologia
13.
Probiotics Antimicrob Proteins ; 13(4): 970-981, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33649898

RESUMO

Infectious mastitis is the major cause of early weaning, depriving infants of breastfeeding benefits. It is associated with an inflammatory condition of the breast and lowered resistance to infection. Drug administration during lactation often being contra-indicated, it is therefore important to consider safe therapeutic alternatives to antibiotic and anti-inflammatory therapies, such as probiotics. In this study, we investigated in vitro the probiotic potential of thirteen Lacticaseibacillus (formerly Lactobacillus) rhamnosus strains isolated from the gut microbiota of breastfed healthy infants. Strains were assessed for their ß-hemolytic activity, their resistance to antibiotics, and their antimicrobial activities against strains of Staphylococcus and Streptococcus, most often involved in women mastitis. Their immunomodulating abilities were also studied using in vitro stimulation of human immune cells. None of the strains exhibited ß-hemolytic activity, and all of them were sensitive to ampicillin, penicillin, tetracycline, rifampicin, erythromycin, chloramphenicol, and imipenem but showed resistance to ceftazidime, trimethoprim/sulfamethoxazole, vancomycin, and cefotaxime, reported to be chromosomally encoded and not inducible or transferable. Four L. rhamnosus strains were selected for their large anti-staphylococcal spectrum: L. rhamnosus VR1-5 and L. rhamnosus VR3-1 inhibiting S. aureus, S. epidermis, and S. warneri and L. rhamnosus CB9-2 and L. rhamnosus CB10-5 exerting antagonistic effect against S. aureus and S. epidermis strains. Antimicrobial compounds released in cell-free supernatant showed proteinaceous nature and were thermoresistant. The immune modulatory analysis of the L. rhamnosus strains revealed two strains with significant anti-inflammatory potential, highlighted by strong induction of IL-10 and a weak pro-Th1 cytokine secretion (IL-12 and IFN-γ). L. rhamnosus CB9-2 combined a large anti-staphylococcal activity spectrum and a promising anti-inflammatory profile. This strain, used individually or in a mixture, can be considered as a probiotic candidate for the management of infectious mastitis during lactation.


Assuntos
Anti-Inflamatórios , Microbioma Gastrointestinal , Lacticaseibacillus rhamnosus , Mastite , Probióticos , Feminino , Humanos , Lactente , Mastite/terapia , Staphylococcus aureus , Staphylococcus epidermidis
14.
Int J Mol Sci ; 23(1)2021 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-35008535

RESUMO

Mitochondrial dysfunctions are implicated in several pathologies, such as metabolic, cardiovascular, respiratory, and neurological diseases, as well as in cancer and aging. These metabolic alterations are usually assessed in human or murine samples by mitochondrial respiratory chain enzymatic assays, by measuring the oxygen consumption of intact mitochondria isolated from tissues, or from cells obtained after physical or enzymatic disruption of the tissues. However, these methodologies do not maintain tissue multicellular organization and cell-cell interactions, known to influence mitochondrial metabolism. Here, we develop an optimal model to measure mitochondrial oxygen consumption in heart and lung tissue samples using the XF24 Extracellular Flux Analyzer (Seahorse) and discuss the advantages and limitations of this technological approach. Our results demonstrate that tissue organization, as well as mitochondrial ultrastructure and respiratory function, are preserved in heart and lung tissues freshly processed or after overnight conservation at 4 °C. Using this method, we confirmed the repeatedly reported obesity-associated mitochondrial dysfunction in the heart and extended it to the lungs. We set up and validated a new strategy to optimally assess mitochondrial function in murine tissues. As such, this method is of great potential interest for monitoring mitochondrial function in cohort samples.


Assuntos
Consumo de Oxigênio/fisiologia , Envelhecimento/fisiologia , Animais , Comunicação Celular/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Respiração Celular/fisiologia , Metabolismo Energético/fisiologia , Coração/fisiologia , Humanos , Pulmão/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/fisiologia , Membranas Mitocondriais/fisiologia , Ratos , Testes de Função Respiratória/métodos
15.
Cells ; 9(9)2020 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-32947881

RESUMO

Alterations in the gut microbiota composition and diversity seem to play a role in the development of chronic diseases, including inflammatory bowel disease (IBD), leading to gut barrier disruption and induction of proinflammatory immune responses. This opens the door for the use of novel health-promoting bacteria. We selected five Parabacteroides distasonis strains isolated from human adult and neonates gut microbiota. We evaluated in vitro their immunomodulation capacities and their ability to reinforce the gut barrier and characterized in vivo their protective effects in an acute murine model of colitis. The in vitro beneficial activities were highly strain dependent: two strains exhibited a potent anti-inflammatory potential and restored the gut barrier while a third strain reinstated the epithelial barrier. While their survival to in vitro gastric conditions was variable, the levels of P. distasonis DNA were higher in the stools of bacteria-treated animals. The strains that were positively scored in vitro displayed a strong ability to rescue mice from colitis. We further showed that two strains primed dendritic cells to induce regulatory T lymphocytes from naïve CD4+ T cells. This study provides better insights on the functionality of commensal bacteria and crucial clues to design live biotherapeutics able to target inflammatory chronic diseases such as IBD.


Assuntos
Bacteroidetes/genética , Bacteroidetes/imunologia , Colite/induzido quimicamente , Colite/microbiologia , Microbioma Gastrointestinal/imunologia , Ácido Trinitrobenzenossulfônico/efeitos adversos , Adulto , Animais , Bacteroidetes/isolamento & purificação , Células CACO-2 , Colite/imunologia , DNA Bacteriano/genética , DNA Bacteriano/metabolismo , Modelos Animais de Doenças , Fezes/microbiologia , Feminino , Humanos , Recém-Nascido , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/microbiologia , Mucosa Intestinal/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase em Tempo Real , Linfócitos T Reguladores/imunologia
16.
Nutrients ; 12(3)2020 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-32244932

RESUMO

Unhealthy lifestyle choices, such as bad eating behaviors and cigarette smoking, have major detrimental impacts on health. However, the inter-relations between obesity and smoking are still not fully understood. We thus developed an experimental model of high-fat diet-fed obese C57BL/6 male mice chronically exposed to cigarette smoke. Our study evaluated for the first time the resulting effects of the combined exposure to unhealthy diet and cigarette smoke on several metabolic, pulmonary, intestinal, and cardiac parameters. We showed that the chronic exposure to cigarette smoke modified the pattern of body fat distribution in favor of the visceral depots in obese mice, impaired the respiratory function, triggered pulmonary inflammation and emphysema, and was associated with gut microbiota dysbiosis, cardiac hypertrophy and myocardial fibrosis.


Assuntos
Exposição Ambiental , Estilo de Vida , Obesidade/etiologia , Fumar/efeitos adversos , Tecido Adiposo/metabolismo , Animais , Biomarcadores , Cardiomegalia/etiologia , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Citocinas/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Metabolismo Energético , Glucose/metabolismo , Homeostase , Humanos , Insulina/metabolismo , Pulmão/fisiopatologia , Masculino , Camundongos , Microbiota , Obesidade/complicações , Obesidade/metabolismo , Especificidade de Órgãos
17.
Sci Rep ; 10(1): 5345, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32210304

RESUMO

Crohn's disease is linked to a decreased diversity in gut microbiota composition as a potential consequence of an impaired anti-microbial response and an altered polarization of T helper cells. Here, we evaluated the immunomodulatory properties of two potential probiotic strains, namely a Bifidobacterium animalis spp. lactis Bl 5764 and a Lactobacillus reuteri Lr 5454 strains. Both strains improved colitis triggered by either 2,4,6-trinitrobenzenesulfonic acid (TNBS) or Citrobacter rodentium infection in mice. Training of dendritic cells (DC) with Lr 5454 efficiently triggered IL-22 secretion and regulatory T cells induction in vitro, while IL-17A production by CD4+ T lymphocytes was stronger when cultured with DCs that were primed with Bl 5764. This strain was sufficient for significantly inducing expression of antimicrobial peptides in vivo through the Crohn's disease predisposing gene encoding for the nucleotide-binding oligomerization domain, containing protein 2 (NOD2). In contrast, NOD2 was dispensable for the impact on antimicrobial peptide expression in mice that were monocolonized with Lr 5454. In conclusion, our work highlights a differential mode of action of two potential probiotic strains that protect mice against colitis, providing the rational for a personalized supportive preventive therapy by probiotics for individuals that are genetically predisposed to Crohn's disease.


Assuntos
Bifidobacterium animalis , Colite/microbiologia , Colite/terapia , Células Dendríticas/fisiologia , Limosilactobacillus reuteri , Probióticos/farmacologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Citrobacter rodentium/patogenicidade , Colite/induzido quimicamente , Colite/patologia , Modelos Animais de Doenças , Infecções por Enterobacteriaceae/microbiologia , Feminino , Microbioma Gastrointestinal , Vida Livre de Germes , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas a Pancreatite/genética , Linfócitos T Auxiliares-Indutores/fisiologia , Linfócitos T Reguladores/fisiologia , Ácido Trinitrobenzenossulfônico/toxicidade
18.
Cell Physiol Biochem ; 53(5): 774-793, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31647207

RESUMO

BACKGROUND/AIMS: Deregulation of the complex interaction among host genetics, gut microbiota and environmental factors on one hand and aberrant immune responses on the other hand, are known to be associated with the development of inflammatory bowel disease. Recent studies provided strong evidence that autophagy plays a key role in the etiology of Crohn's disease (CD). Probiotics may exhibit many therapeutic properties, including anti-inflammatory abilities. While successful results have been obtained in ulcerative colitis patients, probiotics remain inefficient in CD for unknown reason. It remains therefore important to better understand their molecular mechanisms of action. METHODS: The activation of autophagy was examined by stimulating bone marrow-derived dendritic cells by the bacteria, followed by confocal microscopy and western blot analysis. The impact of blocking in vitro autophagy was performed in peripheral blood mononuclear cells using 3-methyl adenine or bafilomycin followed by cytokine secretion measurement by ELISA. The role of autophagy in the anti-inflammatory capacities of the bacterial strains was evaluated in vivo using an acute trinitrobenzene sulfonic acid-induced murine model of colitis. The impact of BMDC was evaluated by adoptive transfer, notably using bone marrow cells derived from autophagy-related 16-like 1-deficient mice. RESULTS: We showed that selected lactobacilli and bifidobacteria are able to induce autophagy activation in BMDCs. Blocking in vitro autophagy abolished the capacity of the strains to induce the release of the anti-inflammatory cytokine interleukin-10, while it exacerbated the secretion of the pro-inflammatory cytokine interleukin-1ß. We confirmed in the TNBS-induced mouse model of colitis that autophagy is involved in the protective capacity of these selected strains, and showed that dendritic cells are involved in this process. CONCLUSION: We propose autophagy as a novel mechanism involved in the regulatory capacities of probiotics.


Assuntos
Autofagia , Bifidobacterium/fisiologia , Lactobacillus/fisiologia , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Proteínas Relacionadas à Autofagia , Células da Medula Óssea/citologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Quimiocinas/genética , Quimiocinas/metabolismo , Colite/induzido quimicamente , Colite/microbiologia , Colite/patologia , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Células Dendríticas/microbiologia , Feminino , Humanos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Macrolídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout
19.
Sci Rep ; 7: 43211, 2017 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-28233848

RESUMO

Gut microbiota dysbiosis plays a central role in the development and perpetuation of chronic inflammation in inflammatory bowel disease (IBD) and therefore is key target for interventions with high quality and functional probiotics. The local production of stable probiotic formulations at limited cost is considered an advantage as it reduces transportation cost and time, thereby increasing the effective period at the consumer side. In the present study, we compared the anti-inflammatory capacities of the Bifidobacterium animalis subsp. lactis (B. lactis) INL1, a probiotic strain isolated in Argentina from human breast milk, with the commercial strain B. animalis subsp. lactis BB12. The impact of spray-drying, a low-cost alternative of bacterial dehydration, on the functionality of both bifidobacteria was also investigated. We showed for both bacteria that the spray-drying process did not impact on bacterial survival nor on their protective capacities against acute and chronic colitis in mice, opening future perspectives for the use of strain INL1 in populations with IBD.


Assuntos
Bifidobacterium animalis/isolamento & purificação , Colite/prevenção & controle , Dessecação/métodos , Leite Humano/microbiologia , Probióticos/administração & dosagem , Probióticos/isolamento & purificação , Tecnologia Farmacêutica/métodos , Animais , Argentina , Técnicas Bacteriológicas/métodos , Bifidobacterium animalis/fisiologia , Modelos Animais de Doenças , Humanos , Camundongos , Viabilidade Microbiana
20.
ISME J ; 10(6): 1456-70, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26613342

RESUMO

Cancer cachexia is a multifactorial syndrome that includes muscle wasting and inflammation. As gut microbes influence host immunity and metabolism, we investigated the role of the gut microbiota in the therapeutic management of cancer and associated cachexia. A community-wide analysis of the caecal microbiome in two mouse models of cancer cachexia (acute leukaemia or subcutaneous transplantation of colon cancer cells) identified common microbial signatures, including decreased Lactobacillus spp. and increased Enterobacteriaceae and Parabacteroides goldsteinii/ASF 519. Building on this information, we administered a synbiotic containing inulin-type fructans and live Lactobacillus reuteri 100-23 to leukaemic mice. This treatment restored the Lactobacillus population and reduced the Enterobacteriaceae levels. It also reduced hepatic cancer cell proliferation, muscle wasting and morbidity, and prolonged survival. Administration of the synbiotic was associated with restoration of the expression of antimicrobial proteins controlling intestinal barrier function and gut immunity markers, but did not impact the portal metabolomics imprinting of energy demand. In summary, this study provided evidence that the development of cancer outside the gut can impact intestinal homeostasis and the gut microbial ecosystem and that a synbiotic intervention, by targeting some alterations of the gut microbiota, confers benefits to the host, prolonging survival and reducing cancer proliferation and cachexia.


Assuntos
Caquexia/microbiologia , Neoplasias do Colo/microbiologia , Microbioma Gastrointestinal/fisiologia , Lactobacillus/fisiologia , Leucemia/microbiologia , Simbióticos/administração & dosagem , Doença Aguda , Animais , Bacteroidetes/efeitos dos fármacos , Bacteroidetes/crescimento & desenvolvimento , Caquexia/terapia , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/terapia , Modelos Animais de Doenças , Enterobacteriaceae/efeitos dos fármacos , Enterobacteriaceae/crescimento & desenvolvimento , Feminino , Frutanos/administração & dosagem , Microbioma Gastrointestinal/efeitos dos fármacos , Homeostase , Inflamação , Intestinos/microbiologia , Inulina/administração & dosagem , Lactobacillus/efeitos dos fármacos , Lactobacillus/crescimento & desenvolvimento , Leucemia/terapia , Metabolômica , Camundongos , Camundongos Endogâmicos BALB C
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...