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










Base de dados
Intervalo de ano de publicação
1.
J Appl Toxicol ; 41(2): 276-290, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32725676

RESUMO

Gut microorganisms are vital for many aspects of human health, and the commensal bacterium Akkermansia muciniphila has repeatedly been identified as a key component of intestinal microbiota. Reductions in A. muciniphila abundance are associated with increased prevalence of metabolic disorders such as obesity and type 2 diabetes. It was recently discovered that administration of A. muciniphila has beneficial effects and that these are not diminished, but rather enhanced after pasteurization. Pasteurized A. muciniphila is proposed for use as a food ingredient, and was therefore subjected to a nonclinical safety assessment, comprising genotoxicity assays (bacterial reverse mutation and in vitro mammalian cell micronucleus tests) and a 90-day toxicity study. For the latter, Han Wistar rats were administered with the vehicle or pasteurized A. muciniphila at doses of 75, 375 or 1500 mg/kg body weight/day (equivalent to 4.8 × 109 , 2.4 × 1010 , or 9.6 × 1010 A. muciniphila cells/kg body weight/day) by oral gavage for 90 consecutive days. The study assessed potential effects on clinical observations (including detailed arena observations and a modified Irwin test), body weight, food and water consumption, clinical pathology, organ weights, and macroscopic and microscopic pathology. The results of both in vitro genotoxicity studies were negative. No test item-related adverse effects were observed in the 90-day study; therefore, 1500 mg/kg body weight/day (the highest dose tested, equivalent to 9.6 × 1010 A. muciniphila cells/kg body weight/day) was established as the no-observed-adverse-effect-level. These results support that pasteurized A. muciniphila is safe for use as a food ingredient.


Assuntos
Akkermansia/crescimento & desenvolvimento , Akkermansia/efeitos da radiação , Suplementos Nutricionais/toxicidade , Inocuidade dos Alimentos , Microbioma Gastrointestinal/efeitos da radiação , Pasteurização , Animais , Humanos , Masculino , Modelos Animais , Ratos , Ratos Wistar
2.
Proc Natl Acad Sci U S A ; 117(42): 26374-26381, 2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-33020286

RESUMO

Mechanistic understanding of the factors that govern host tropism remains incompletely understood for most pathogens. Brucella species, which are capable of infecting a wide range of hosts, offer a useful avenue to address this question. We hypothesized that metabolic fine-tuning to intrahost niches is likely an underappreciated axis underlying pathogens' ability to infect new hosts and tropism. In this work, we compared the central metabolism of seven Brucella species by stable isotopic labeling and genetics. We identified two functionally distinct groups, one overlapping with the classical zoonotic species of domestic livestock that exclusively use the pentose phosphate pathway (PPP) for hexose catabolism, whereas species from the second group use mostly the Entner-Doudoroff pathway (EDP). We demonstrated that the metabolic dichotomy among Brucellae emerged after the acquisition of two independent EDP-inactivating mutations in all classical zoonotic species. We then examined the pathogenicity of key metabolic mutants in mice and confirmed that this trait is tied to virulence. Altogether, our data are consistent with the hypothesis that the PPP has been incrementally selected over the EDP in parallel to Brucella adaptation to domestic livestock.


Assuntos
Brucella/genética , Brucella/metabolismo , Via de Pentose Fosfato/genética , Adaptação Biológica/genética , Animais , Zoonoses Bacterianas/genética , Evolução Biológica , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Via de Pentose Fosfato/fisiologia , Fenótipo , Virulência
3.
Am J Physiol Endocrinol Metab ; 318(4): E480-E491, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31961709

RESUMO

Probiotic bacteria can protect from ovariectomy (ovx)-induced bone loss in mice. Akkermansia muciniphila is considered to have probiotic potential due to its beneficial effect on obesity and insulin resistance. The purpose of the present study was to determine if treatment with pasteurized Akkermansia muciniphila (pAkk) could prevent ovx-induced bone loss. Mice were treated with vehicle or pAkk for 4 wk, starting 3 days before ovx or sham surgery. Treatment with pAkk reduced fat mass accumulation confirming earlier findings. However, treatment with pAkk decreased trabecular and cortical bone mass in femur and vertebra of gonadal intact mice and did not protect from ovx-induced bone loss. Treatment with pAkk increased serum parathyroid hormone (PTH) levels and increased expression of the calcium transporter Trpv5 in kidney suggesting increased reabsorption of calcium in the kidneys. Serum amyloid A 3 (SAA3) can suppress bone formation and mediate the effects of PTH on bone resorption and bone loss in mice and treatment with pAkk increased serum levels of SAA3 and gene expression of Saa3 in colon. Moreover, regulatory T cells can be protective of bone and pAkk-treated mice had decreased number of regulatory T cells in mesenteric lymph nodes and bone marrow. In conclusion, treatment with pAkk protected from ovx-induced fat mass gain but not from bone loss and reduced bone mass in gonadal intact mice. Our findings with pAkk differ from some probiotics that have been shown to protect bone mass, demonstrating that not all prebiotic and probiotic factors have the same effect on bone.


Assuntos
Tecido Adiposo/crescimento & desenvolvimento , Microbioma Gastrointestinal/fisiologia , Osteoporose/metabolismo , Probióticos/farmacologia , Verrucomicrobia/metabolismo , Tecido Adiposo/metabolismo , Akkermansia , Animais , Canais de Cálcio/metabolismo , Colo/efeitos dos fármacos , Colo/microbiologia , Feminino , Fêmur/efeitos dos fármacos , Linfonodos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Ovariectomia , Hormônio Paratireóideo/metabolismo , Pasteurização , Proteína Amiloide A Sérica/metabolismo , Coluna Vertebral/efeitos dos fármacos , Linfócitos T Reguladores , Canais de Cátion TRPV/metabolismo
4.
Nat Med ; 25(7): 1096-1103, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31263284

RESUMO

Metabolic syndrome is characterized by a constellation of comorbidities that predispose individuals to an increased risk of developing cardiovascular pathologies as well as type 2 diabetes mellitus1. The gut microbiota is a new key contributor involved in the onset of obesity-related disorders2. In humans, studies have provided evidence for a negative correlation between Akkermansia muciniphila abundance and overweight, obesity, untreated type 2 diabetes mellitus or hypertension3-8. Since the administration of A. muciniphila has never been investigated in humans, we conducted a randomized, double-blind, placebo-controlled pilot study in overweight/obese insulin-resistant volunteers; 40 were enrolled and 32 completed the trial. The primary end points were safety, tolerability and metabolic parameters (that is, insulin resistance, circulating lipids, visceral adiposity and body mass). Secondary outcomes were gut barrier function (that is, plasma lipopolysaccharides) and gut microbiota composition. In this single-center study, we demonstrated that daily oral supplementation of 1010 A. muciniphila bacteria either live or pasteurized for three months was safe and well tolerated. Compared to placebo, pasteurized A. muciniphila improved insulin sensitivity (+28.62 ± 7.02%, P = 0.002), and reduced insulinemia (-34.08 ± 7.12%, P = 0.006) and plasma total cholesterol (-8.68 ± 2.38%, P = 0.02). Pasteurized A. muciniphila supplementation slightly decreased body weight (-2.27 ± 0.92 kg, P = 0.091) compared to the placebo group, and fat mass (-1.37 ± 0.82 kg, P = 0.092) and hip circumference (-2.63 ± 1.14 cm, P = 0.091) compared to baseline. After three months of supplementation, A. muciniphila reduced the levels of the relevant blood markers for liver dysfunction and inflammation while the overall gut microbiome structure was unaffected. In conclusion, this proof-of-concept study (clinical trial no. NCT02637115 ) shows that the intervention was safe and well tolerated and that supplementation with A. muciniphila improves several metabolic parameters.


Assuntos
Suplementos Nutricionais , Obesidade/dietoterapia , Sobrepeso/dietoterapia , Verrucomicrobia , Adulto , Idoso , Método Duplo-Cego , Fezes/microbiologia , Microbioma Gastrointestinal , Humanos , Resistência à Insulina , Pessoa de Meia-Idade , Obesidade/metabolismo , Obesidade/microbiologia , Sobrepeso/metabolismo , Sobrepeso/microbiologia , Projetos Piloto
5.
Nat Commun ; 10(1): 457, 2019 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-30692526

RESUMO

Variations in N-acylethanolamines (NAE) levels are associated with obesity and metabolic comorbidities. Their role in the gut remains unclear. Therefore, we generated a mouse model of inducible intestinal epithelial cell (IEC)-specific deletion of N-acylphosphatidylethanolamine phospholipase D (NAPE-PLD), a key enzyme involved in NAE biosynthesis (Napepld∆IEC). We discovered that Napepld∆IEC mice are hyperphagic upon first high-fat diet (HFD) exposure, and develop exacerbated obesity and steatosis. These mice display hypothalamic Pomc neurons dysfunctions and alterations in intestinal and plasma NAE and 2-acylglycerols. After long-term HFD, Napepld∆IEC mice present reduced energy expenditure. The increased steatosis is associated with higher gut and liver lipid absorption. Napepld∆IEC mice display altered gut microbiota. Akkermansia muciniphila administration partly counteracts the IEC NAPE-PLD deletion effects. In conclusion, intestinal NAPE-PLD is a key sensor in nutritional adaptation to fat intake, gut-to-brain axis and energy homeostasis and thereby constitutes a novel target to tackle obesity and related disorders.


Assuntos
Gorduras na Dieta/metabolismo , Fígado Gorduroso/metabolismo , Mucosa Intestinal/metabolismo , Obesidade/metabolismo , Fosfatidiletanolaminas/metabolismo , Fosfolipase D/metabolismo , Adaptação Fisiológica , Animais , Dieta Hiperlipídica/efeitos adversos , Fígado Gorduroso/etiologia , Microbioma Gastrointestinal/fisiologia , Homeostase , Mucosa Intestinal/microbiologia , Metabolismo dos Lipídeos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Obesidade/etiologia
6.
Oncotarget ; 9(26): 18224-18238, 2018 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-29719601

RESUMO

Intestinal disorders often occur in cancer patients, in association with body weight loss, and this alteration is commonly attributed to the chemotherapy. Here, using a mouse model of cancer cachexia induced by ectopic transplantation of C26 cancer cells, we discovered a profound alteration in the gut functions (gut permeability, epithelial turnover, gut immunity, microbial dysbiosis) independently of any chemotherapy. These alterations occurred independently of anorexia and were driven by interleukin 6. Gut dysfunction was found to be resistant to treatments with an anti-inflammatory bacterium (Faecalibacterium prausnitzii) or with gut peptides involved in intestinal cell renewal (teduglutide, a glucagon-like peptide 2 analogue). The translational value of our findings was evaluated in 152 colorectal and lung cancer patients with or without cachexia. The serum level of the lipopolysaccharide-binding protein, often presented as a reflection of the bacterial antigen load, was not only increased in cachectic mice and cancer patients, but also strongly correlated with the serum IL-6 level and predictive of death and cachexia occurrence in these patients. Altogether, our data highlight profound alterations of the intestinal homeostasis in cancer cachexia occurring independently of any chemotherapy and food intake reduction, with potential relevance in humans. In addition, we point out the lipopolysaccharide-binding protein as a new biomarker of cancer cachexia related to gut dysbiosis.

7.
Sci Total Environ ; 627: 1018-1038, 2018 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-29426121

RESUMO

Plants, animals and humans, are colonized by microorganisms (microbiota) and transiently exposed to countless others. The microbiota affects the development and function of essentially all organ systems, and contributes to adaptation and evolution, while protecting against pathogenic microorganisms and toxins. Genetics and lifestyle factors, including diet, antibiotics and other drugs, and exposure to the natural environment, affect the composition of the microbiota, which influences host health through modulation of interrelated physiological systems. These include immune system development and regulation, metabolic and endocrine pathways, brain function and epigenetic modification of the genome. Importantly, parental microbiotas have transgenerational impacts on the health of progeny. Humans, animals and plants share similar relationships with microbes. Research paradigms from humans and other mammals, amphibians, insects, planktonic crustaceans and plants demonstrate the influence of environmental microbial ecosystems on the microbiota and health of organisms, and indicate links between environmental and internal microbial diversity and good health. Therefore, overlapping compositions, and interconnected roles of microbes in human, animal and plant health should be considered within the broader context of terrestrial and aquatic microbial ecosystems that are challenged by the human lifestyle and by agricultural and industrial activities. Here, we propose research priorities and organizational, educational and administrative measures that will help to identify safe microbe-associated health-promoting modalities and practices. In the spirit of an expanding version of "One health" that includes environmental health and its relation to human cultures and habits (EcoHealth), we urge that the lifestyle-microbiota-human health nexus be taken into account in societal decision making.


Assuntos
Ecossistema , Microbiologia Ambiental , Atividades Humanas , Animais , Monitoramento Ambiental , Humanos , Estilo de Vida , Microbiota , Plantas
8.
Gut ; 67(2): 271-283, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28377388

RESUMO

OBJECTIVE: To investigate the beneficial role of prebiotics on endothelial dysfunction, an early key marker of cardiovascular diseases, in an original mouse model linking steatosis and endothelial dysfunction. DESIGN: We examined the contribution of the gut microbiota to vascular dysfunction observed in apolipoprotein E knockout (Apoe-/-) mice fed an n-3 polyunsaturated fatty acid (PUFA)-depleted diet for 12 weeks with or without inulin-type fructans (ITFs) supplementation for the last 15 days. Mesenteric and carotid arteries were isolated to evaluate endothelium-dependent relaxation ex vivo. Caecal microbiota composition (Illumina Sequencing of the 16S rRNA gene) and key pathways/mediators involved in the control of vascular function, including bile acid (BA) profiling, gut and liver key gene expression, nitric oxide and gut hormones production were also assessed. RESULTS: ITF supplementation totally reverses endothelial dysfunction in mesenteric and carotid arteries of n-3 PUFA-depleted Apoe-/- mice via activation of the nitric oxide (NO) synthase/NO pathway. Gut microbiota changes induced by prebiotic treatment consist in increased NO-producing bacteria, replenishment of abundance in Akkermansia and decreased abundance in bacterial taxa involved in secondary BA synthesis. Changes in gut and liver gene expression also occur upon ITFs suggesting increased glucagon-like peptide 1 production and BA turnover as drivers of endothelium function preservation. CONCLUSIONS: We demonstrate for the first time that ITF improve endothelial dysfunction, implicating a short-term adaptation of both gut microbiota and key gut peptides. If confirmed in humans, prebiotics could be proposed as a novel approach in the prevention of metabolic disorders-related cardiovascular diseases.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Frutanos/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Prebióticos , Aminopeptidases/genética , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Bactérias/efeitos dos fármacos , Ácidos e Sais Biliares/biossíntese , Ácidos e Sais Biliares/sangue , Artérias Carótidas/fisiologia , Ceco/microbiologia , Suplementos Nutricionais , Modelos Animais de Doenças , Ácidos Graxos Ômega-3/administração & dosagem , Ácidos Graxos Ômega-3/deficiência , Expressão Gênica/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/biossíntese , Masculino , Artérias Mesentéricas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Neurotensina/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Proglucagon/genética , Simportadores/genética , Vasodilatação
9.
Am J Physiol Endocrinol Metab ; 314(4): E334-E352, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28874357

RESUMO

Increasing evidence suggests that polyphenols have a significant potential in the prevention and treatment of risk factors associated with metabolic syndrome. The objective of this study was to assess the metabolic outcomes of two polyphenol-containing extracts from cinnamon bark (CBE) and grape pomace (GPE) on C57BL/6J mice fed a high-fat diet (HFD) for 8 wk. Both CBE and GPE were able to decrease fat mass gain and adipose tissue inflammation in mice fed a HFD without reducing food intake. This was associated with reduced liver steatosis and lower plasma nonesterified fatty acid levels. We also observed a beneficial effect on glucose homeostasis, as evidenced by an improved glucose tolerance and a lower insulin resistance index. These ameliorations of the overall metabolic profile were associated with a significant impact on the microbial composition, which was more profound for the GPE than for the CBE. At the genus level, Peptococcus were decreased in the CBE group. In the GPE-treated group, several key genera that have been previously found to be linked with HFD, metabolic effects, and gut barrier integrity were affected: we observed a decrease of Desulfovibrio, Lactococcus, whereas Allobaculum and Roseburia were increased. In addition, the expression of several antimicrobial peptides and tight junction proteins was increased in response to both CBE and GPE supplementation, indicating an improvement of the gut barrier function. Collectively, these data suggest that CBE and GPE can ameliorate the overall metabolic profile of mice on a high-fat diet, partly by acting on the gut microbiota.


Assuntos
Cinnamomum zeylanicum/química , Microbioma Gastrointestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Doenças Metabólicas/prevenção & controle , Extratos Vegetais/farmacologia , Vitis/química , Animais , Biomarcadores/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/microbiologia , Diabetes Mellitus Experimental/prevenção & controle , Dieta Hiperlipídica/efeitos adversos , Fígado Gorduroso/metabolismo , Fígado Gorduroso/microbiologia , Fígado Gorduroso/prevenção & controle , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Masculino , Doenças Metabólicas/etiologia , Doenças Metabólicas/metabolismo , Doenças Metabólicas/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Obesidade/microbiologia , Obesidade/prevenção & controle , Permeabilidade , Extratos Vegetais/uso terapêutico
10.
Gut ; 67(8): 1445-1453, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29269438

RESUMO

OBJECTIVE: Intestinal microbiota is implicated in the pathogenesis of autoimmune type 1 diabetes in humans and in non-obese diabetic (NOD) mice, but evidence on its causality and on the role of individual microbiota members is limited. We investigated if different diabetes incidence in two NOD colonies was due to microbiota differences and aimed to identify individual microbiota members with potential significance. DESIGN: We profiled intestinal microbiota between two NOD mouse colonies showing high or low diabetes incidence by 16S ribosomal RNA gene sequencing and colonised the high-incidence colony with the microbiota of the low-incidence colony. Based on unaltered incidence, we identified a few taxa which were not effectively transferred and thereafter, transferred experimentally one of these to test its potential significance. RESULTS: Although the high-incidence colony adopted most microbial taxa present in the low-incidence colony, diabetes incidence remained unaltered. Among the few taxa which were not transferred, Akkermansia muciniphila was identified. As A. muciniphila abundancy is inversely correlated to the risk of developing type 1 diabetes-related autoantibodies, we transferred A. muciniphila experimentally to the high-incidence colony. A. muciniphila transfer promoted mucus production and increased expression of antimicrobial peptide Reg3γ, outcompeted Ruminococcus torques from the microbiota, lowered serum endotoxin levels and islet toll-like receptor expression, promoted regulatory immunity and delayed diabetes development. CONCLUSION: Transfer of the whole microbiota may not reduce diabetes incidence despite a major change in gut microbiota, but single symbionts such as A. muciniphila with beneficial metabolic and immune signalling effects may reduce diabetes incidence when administered as a probiotic.


Assuntos
Diabetes Mellitus Tipo 1/microbiologia , Microbioma Gastrointestinal/fisiologia , Verrucomicrobia , Animais , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/metabolismo , Interleucina-10/metabolismo , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Endogâmicos NOD , Linfócitos T Reguladores , Receptores Toll-Like/metabolismo
11.
Endocr Dev ; 32: 139-164, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28898875

RESUMO

Gut bacteria exert a variety of metabolic functions unavailable to the host and are increasingly seen as a virtual organ located inside our gastrointestinal tract. Scattered in our intestinal epithelium, enteroendocrine cells (EECs) regulate several aspects of the host's physiology and translate signals coming from the gut microbiota through their hormonal secretions. In this chapter, we will assess the interplay between the gut microbiota and EEC and its consequences for the physiology of the host. We will first describe alterations of different populations of EEC in germ-free animals. The role of mediators of this interaction, such as microbial metabolites and their receptors will also be discussed. Finally, different strategies harnessing host-microbe crosstalk for therapeutic purposes will be presented with an emphasis on obesity and related disorders.


Assuntos
Células Enteroendócrinas/fisiologia , Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Interações Hospedeiro-Patógeno/fisiologia , Redes e Vias Metabólicas/fisiologia , Animais , Células Enteroendócrinas/metabolismo , Células Enteroendócrinas/microbiologia , Humanos , Obesidade/metabolismo , Obesidade/microbiologia
12.
Microbiol Spectr ; 5(3)2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28597812

RESUMO

Malnutrition is the cause of major public health concerns worldwide. On the one hand, obesity and associated pathologies (also known as the metabolic syndrome) affect more than 10% of the world population. Such pathologies might arise from an elevated inflammatory tone. We have discovered that the inflammatory properties of high-fat diets were linked to the translocation of lipopolysaccharide (LPS). We proposed a mechanism associating the gut microbiota with the onset of insulin resistance and low-grade inflammation, a phenomenon that we called "metabolic endotoxemia." We and others have shown that bacteria as well as host-derived immune-related elements control microbial communities and eventually contribute to the phenotype observed during diet-induced obesity, diabetes, and metabolic inflammation. On the other hand, undernutrition is one of the leading causes of death in children. A diet poor in energy and/or nutrients causes incomplete development of the gut microbiota and may profoundly affect energy absorption, initiating stunted growth, edema, and diarrhea. In this review, we discuss how changes in microbiota composition are associated with obesity and undernutrition. We also highlight that opposite consequences exist in terms of energy absorption from the diet (obesity versus undernutrition), but interestingly the two situations share similar defects in term of diversity, functionality, and inflammatory potential.


Assuntos
Metabolismo Energético/fisiologia , Microbioma Gastrointestinal/fisiologia , Distúrbios Nutricionais/terapia , Obesidade/microbiologia , Probióticos/uso terapêutico , Animais , Diabetes Mellitus , Dieta Hiperlipídica/efeitos adversos , Endotoxemia , Microbioma Gastrointestinal/genética , Vida Livre de Germes , Humanos , Inflamação/metabolismo , Resistência à Insulina , Lipopolissacarídeos/metabolismo , Camundongos , Modelos Biológicos
13.
Front Microbiol ; 8: 1088, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28659902

RESUMO

Erythritol is the preferential carbon source for most brucellae, a group of facultative intracellular bacteria that cause a worldwide zoonosis. Since this polyol is abundant in genital organs of ruminants and swine, it is widely accepted that erythritol accounts at least in part for the characteristic genital tropism of brucellae. Nevertheless, proof of erythritol availability and essentiality during Brucella intracellular multiplication has remained elusive. To investigate this relationship, we compared ΔeryH (erythritol-sensitive and thus predicted to be attenuated if erythritol is present), ΔeryA (erythritol-tolerant but showing reduced growth if erythritol is a crucial nutrient) and wild type B. abortus in various infection models. This reporting system indicated that erythritol was available but not required for B. abortus multiplication in bovine trophoblasts. However, mice and humans have been considered to lack erythritol, and we found that it was available but not required for B. abortus multiplication in human and murine trophoblastic and macrophage-like cells, and in mouse spleen and conceptus (fetus, placenta and envelopes). Using this animal model, we found that B. abortus infected cells and tissues contained aldose reductase, an enzyme that can account for the production of erythritol from pentose cycle precursors.

15.
Gut ; 66(4): 620-632, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27196572

RESUMO

OBJECTIVE: To examine the role of hepatocyte myeloid differentiation primary-response gene 88 (MyD88) on glucose and lipid metabolism. DESIGN: To study the impact of the innate immune system at the level of the hepatocyte and metabolism, we generated mice harbouring hepatocyte-specific deletion of MyD88. We investigated the impact of the deletion on metabolism by feeding mice with a normal control diet or a high-fat diet for 8 weeks. We evaluated body weight, fat mass gain (using time-domain nuclear magnetic resonance), glucose metabolism and energy homeostasis (using metabolic chambers). We performed microarrays and quantitative PCRs in the liver. In addition, we investigated the gut microbiota composition, bile acid profile and both liver and plasma metabolome. We analysed the expression pattern of genes in the liver of obese humans developing non-alcoholic steatohepatitis (NASH). RESULTS: Hepatocyte-specific deletion of MyD88 predisposes to glucose intolerance, inflammation and hepatic insulin resistance independently of body weight and adiposity. These phenotypic differences were partially attributed to differences in gene expression, transcriptional factor activity (ie, peroxisome proliferator activator receptor-α, farnesoid X receptor (FXR), liver X receptors and STAT3) and bile acid profiles involved in glucose, lipid metabolism and inflammation. In addition to these alterations, the genetic deletion of MyD88 in hepatocytes changes the gut microbiota composition and their metabolomes, resembling those observed during diet-induced obesity. Finally, obese humans with NASH displayed a decreased expression of different cytochromes P450 involved in bioactive lipid synthesis. CONCLUSIONS: Our study identifies a new link between innate immunity and hepatic synthesis of bile acids and bioactive lipids. This dialogue appears to be involved in the susceptibility to alterations associated with obesity such as type 2 diabetes and NASH, both in mice and humans.


Assuntos
Ácidos e Sais Biliares/metabolismo , Microbioma Gastrointestinal/genética , Glucose/metabolismo , Hepatócitos/metabolismo , Metabolismo dos Lipídeos/genética , Metaboloma/genética , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Adiposidade , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Peso Corporal , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Dieta Hiperlipídica , Expressão Gênica , Humanos , Imunidade Inata/genética , Resistência à Insulina/genética , Fígado/metabolismo , Receptores X do Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/imunologia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Obesidade/genética , Obesidade/metabolismo , PPAR alfa/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Fator de Transcrição STAT3/metabolismo
16.
Nat Med ; 23(1): 107-113, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27892954

RESUMO

Obesity and type 2 diabetes are associated with low-grade inflammation and specific changes in gut microbiota composition. We previously demonstrated that administration of Akkermansia muciniphila to mice prevents the development of obesity and associated complications. However, the underlying mechanisms of this protective effect remain unclear. Moreover, the sensitivity of A. muciniphila to oxygen and the presence of animal-derived compounds in its growth medium currently limit the development of translational approaches for human medicine. We have addressed these issues here by showing that A. muciniphila retains its efficacy when grown on a synthetic medium compatible with human administration. Unexpectedly, we discovered that pasteurization of A. muciniphila enhanced its capacity to reduce fat mass development, insulin resistance and dyslipidemia in mice. These improvements were notably associated with a modulation of the host urinary metabolomics profile and intestinal energy absorption. We demonstrated that Amuc_1100, a specific protein isolated from the outer membrane of A. muciniphila, interacts with Toll-like receptor 2, is stable at temperatures used for pasteurization, improves the gut barrier and partly recapitulates the beneficial effects of the bacterium. Finally, we showed that administration of live or pasteurized A. muciniphila grown on the synthetic medium is safe in humans. These findings provide support for the use of different preparations of A. muciniphila as therapeutic options to target human obesity and associated disorders.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Glicemia/efeitos dos fármacos , Diabetes Mellitus Tipo 2/metabolismo , Dislipidemias/metabolismo , Proteínas de Membrana/farmacologia , Obesidade/metabolismo , Receptor 2 Toll-Like/efeitos dos fármacos , Verrucomicrobia , Adulto , Animais , Glicemia/metabolismo , Western Blotting , Cromatografia Líquida , Modelos Animais de Doenças , Feminino , Humanos , Resistência à Insulina , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Masculino , Síndrome Metabólica/metabolismo , Camundongos Obesos , Pessoa de Meia-Idade , Receptor 2 Toll-Like/metabolismo
17.
Nat Rev Endocrinol ; 12(3): 133-43, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26678807

RESUMO

Various metabolic disorders are associated with changes in inflammatory tone. Among the latest advances in the metabolism field, the discovery that gut microorganisms have a major role in host metabolism has revealed the possibility of a plethora of associations between gut bacteria and numerous diseases. However, to date, few mechanisms have been clearly established. Accumulating evidence indicates that the endocannabinoid system and related bioactive lipids strongly contribute to several physiological processes and are a characteristic of obesity, type 2 diabetes mellitus and inflammation. In this Review, we briefly define the gut microbiota as well as the endocannabinoid system and associated bioactive lipids. We discuss existing literature regarding interactions between gut microorganisms and the endocannabinoid system, focusing specifically on the triad of adipose tissue, gut bacteria and the endocannabinoid system in the context of obesity and the development of fat mass. We highlight gut-barrier function by discussing the role of specific factors considered to be putative 'gate keepers' or 'gate openers', and their role in the gut microbiota-endocannabinoid system axis. Finally, we briefly discuss data related to the different pharmacological strategies currently used to target the endocannabinoid system, in the context of cardiometabolic disorders and intestinal inflammation.


Assuntos
Endocanabinoides/metabolismo , Endocanabinoides/fisiologia , Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Animais , Microbioma Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/efeitos dos fármacos , Humanos , Receptores de Canabinoides/efeitos dos fármacos
18.
Nat Commun ; 6: 6495, 2015 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-25757720

RESUMO

Obesity is a pandemic disease associated with many metabolic alterations and involves several organs and systems. The endocannabinoid system (ECS) appears to be a key regulator of energy homeostasis and metabolism. Here we show that specific deletion of the ECS synthesizing enzyme, NAPE-PLD, in adipocytes induces obesity, glucose intolerance, adipose tissue inflammation and altered lipid metabolism. We report that Napepld-deleted mice present an altered browning programme and are less responsive to cold-induced browning, highlighting the essential role of NAPE-PLD in regulating energy homeostasis and metabolism in the physiological state. Our results indicate that these alterations are mediated by a shift in gut microbiota composition that can partially transfer the phenotype to germ-free mice. Together, our findings uncover a role of adipose tissue NAPE-PLD on whole-body metabolism and provide support for targeting NAPE-PLD-derived bioactive lipids to treat obesity and related metabolic disorders.


Assuntos
Tecido Adiposo Marrom/metabolismo , Microbioma Gastrointestinal/fisiologia , Intolerância à Glucose/metabolismo , Obesidade/metabolismo , Fosfolipase D/genética , Tecido Adiposo Marrom/patologia , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/patologia , Animais , Distribuição da Gordura Corporal , Temperatura Baixa , Endocanabinoides/metabolismo , Metabolismo Energético/fisiologia , Expressão Gênica , Intolerância à Glucose/genética , Intolerância à Glucose/microbiologia , Intolerância à Glucose/patologia , Inflamação , Masculino , Camundongos , Camundongos Knockout , Obesidade/genética , Obesidade/microbiologia , Obesidade/patologia , Fosfolipase D/deficiência
19.
Nat Commun ; 5: 5648, 2014 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-25476696

RESUMO

Obesity is associated with a cluster of metabolic disorders, low-grade inflammation and altered gut microbiota. Whether host metabolism is controlled by intestinal innate immune system and the gut microbiota is unknown. Here we report that inducible intestinal epithelial cell-specific deletion of MyD88 partially protects against diet-induced obesity, diabetes and inflammation. This is associated with increased energy expenditure, an improved glucose homeostasis, reduced hepatic steatosis, fat mass and inflammation. Protection is transferred following gut microbiota transplantation to germ-free recipients. We also demonstrate that intestinal epithelial MyD88 deletion increases anti-inflammatory endocannabinoids, restores antimicrobial peptides production and increases intestinal regulatory T cells during diet-induced obesity. Targeting MyD88 after the onset of obesity reduces fat mass and inflammation. Our work thus identifies intestinal epithelial MyD88 as a sensor changing host metabolism according to the nutritional status and we show that targeting intestinal epithelial MyD88 constitutes a putative therapeutic target for obesity and related disorders.


Assuntos
Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Obesidade/metabolismo , Animais , Metabolismo Energético , Feminino , Deleção de Genes , Glucose/metabolismo , Humanos , Intestinos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Estado Nutricional , Obesidade/genética , Obesidade/prevenção & controle
20.
Cell Host Microbe ; 14(5): 582-90, 2013 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-24237703

RESUMO

Gut microbiota contribute to host metabolic efficiency by increasing energy availability through the fermentation of dietary fiber and production of short-chain fatty acids (SCFAs) in the colon. SCFAs are proposed to stimulate secretion of the proglucagon (Gcg)-derived incretin hormone GLP-1, which stimulates insulin secretion (incretin response) and inhibits gastric emptying. We find that germ-free (GF) and antibiotic-treated mice, which have severely reduced SCFA levels, have increased basal GLP-1 levels in the plasma and increased Gcg expression in the colon. Increasing energy supply, either through colonization with polysaccharide-fermenting bacteria or through diet, suppressed colonic Gcg expression in GF mice. Increased GLP-1 levels in GF mice did not improve the incretin response but instead slowed intestinal transit. Thus, microbiota regulate the basal levels of GLP-1, and increasing these levels may be an adaptive response to insufficient energy availability in the colon that slows intestinal transit and allows for greater nutrient absorption.


Assuntos
Bactérias/metabolismo , Colo/microbiologia , Fibras na Dieta/metabolismo , Trânsito Gastrointestinal , Animais , Metabolismo dos Carboidratos , Ácidos Graxos Voláteis/metabolismo , Vida Livre de Germes , Peptídeo 1 Semelhante ao Glucagon/sangue , Camundongos , Proglucagon/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...