Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
2.
Nat Immunol ; 18(5): 541-551, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28288099

RESUMO

Inflammatory bowel diseases involve the dynamic interaction of host genetics, the microbiome and inflammatory responses. Here we found lower expression of NLRP12 (which encodes a negative regulator of innate immunity) in human ulcerative colitis, by comparing monozygotic twins and other patient cohorts. In parallel, Nlrp12 deficiency in mice caused increased basal colonic inflammation, which led to a less-diverse microbiome and loss of protective gut commensal strains (of the family Lachnospiraceae) and a greater abundance of colitogenic strains (of the family Erysipelotrichaceae). Dysbiosis and susceptibility to colitis associated with Nlrp12 deficency were reversed equally by treatment with antibodies targeting inflammatory cytokines and by the administration of beneficial commensal Lachnospiraceae isolates. Fecal transplants from mice reared in specific-pathogen-free conditions into germ-free Nlrp12-deficient mice showed that NLRP12 and the microbiome each contributed to immunological signaling that culminated in colon inflammation. These findings reveal a feed-forward loop in which NLRP12 promotes specific commensals that can reverse gut inflammation, while cytokine blockade during NLRP12 deficiency can reverse dysbiosis.


Assuntos
Clostridiales/fisiologia , Colite Ulcerativa/imunologia , Colo/fisiologia , Firmicutes/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Microbiota , RNA Ribossômico 16S/análise , Animais , Biodiversidade , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/microbiologia , Colo/microbiologia , Sulfato de Dextrana , Fezes/microbiologia , Interação Gene-Ambiente , Humanos , Imunidade Inata/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microbiota/genética , Microbiota/imunologia , Simbiose , Gêmeos Monozigóticos
4.
Mol Pharm ; 15(12): 5454-5467, 2018 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-30372084

RESUMO

The goal of this project was to explore and to statistically evaluate the responsible gastrointestinal (GI) factors that are significant factors in explaining the systemic exposure of ibuprofen, between and within human subjects. In a previous study, we determined the solution and total concentrations of ibuprofen as a function of time in aspirated GI fluids, after oral administration of an 800 mg IR tablet (reference standard) of ibuprofen to 20 healthy volunteers in fasted state conditions. In addition, we determined luminal pH and motility pressure recordings that were simultaneously monitored along the GI tract. Blood samples were taken to determine ibuprofen plasma levels. In this work, an in-depth statistical and pharmacokinetic analysis was performed to explain which underlying GI variables are determining the systemic concentrations of ibuprofen between (inter-) and within (intra-) subjects. In addition, the obtained plasma profiles were deconvoluted to link the fraction absorbed with the fraction dissolved. Multiple linear regressions were performed to explain and quantitatively express the impact of underlying GI physiology on systemic exposure of the drug (in terms of plasma Cmax/AUC and plasma Tmax). The exploratory analysis of the correlation between plasma Cmax/AUC and the time to the first phase III contractions postdose (TMMC-III) explains ∼40% of the variability in plasma Cmax for all fasted state subjects. We have experimentally shown that the in vivo intestinal dissolution of ibuprofen is dependent upon physiological variables like, in this case, pH and postdose phase III contractions. For the first time, this work presents a thorough statistical analysis explaining how the GI behavior of an ionized drug can explain the systemic exposure of the drug based on the individual profiles of participating subjects. This creates a scientifically based and rational framework that emphasizes the importance of including pH and motility in a predictive in vivo dissolution methodology to forecast the in vivo performance of a drug product. Moreover, as no extensive first-pass metabolism is considered for ibuprofen, this study demonstrates how intraluminal drug behavior is reflecting the systemic exposure of a drug.


Assuntos
Liberação Controlada de Fármacos , Jejum/fisiologia , Absorção Gastrointestinal/fisiologia , Trato Gastrointestinal/fisiologia , Ibuprofeno/farmacocinética , Administração Oral , Adulto , Área Sob a Curva , Disponibilidade Biológica , Variação Biológica Individual , Variação Biológica da População/fisiologia , Conjuntos de Dados como Assunto , Feminino , Voluntários Saudáveis , Humanos , Concentração de Íons de Hidrogênio , Ibuprofeno/administração & dosagem , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Solubilidade , Comprimidos , Adulto Jovem
5.
Mol Pharm ; 15(12): 5468-5478, 2018 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-30417648

RESUMO

Exploring the intraluminal behavior of an oral drug product in the human gastrointestinal (GI) tract remains challenging. Many in vivo techniques are available to investigate the impact of GI physiology on oral drug behavior in fasting state conditions. However, little is known about the intraluminal behavior of a drug in postprandial conditions. In a previous report, we described the mean solution and total concentrations of ibuprofen after oral administration of an immediate-release (IR) tablet in fed state conditions. In parallel, blood samples were taken to assess systemic concentrations. The purpose of this work was to statistically evaluate the impact of GI physiology (e.g., pH, contractile events) within and between individuals (intra and intersubject variability) for a total of 17 healthy subjects. In addition, a pharmacokinetic (PK) analysis was performed by noncompartmental analysis, and PK parameters were correlated with underlying physiological factors (pH, time to phase III contractions postdose) and study parameters (e.g., ingested amount of calories, coadministered water). Moreover, individual plasma profiles were deconvoluted to assess the fraction absorbed as a function of time, demonstrating the link between intraluminal and systemic behavior of the drug. The results demonstrated that the in vivo dissolution of ibuprofen depends on the present gastric pH and motility events at the time of administration. Both intraluminal factors were responsible for explaining 63% of plasma Cmax variability among all individuals. For the first time, an in-depth analysis was performed on a large data set derived from an aspiration/motility study, quantifying the impact of physiology on systemic behavior of an orally administered drug product in fed state conditions. The data obtained from this study will help us to develop an in vitro biorelevant dissolution approach and optimize in silico tools in order to predict the in vivo performance of orally administered drug products, especially in fed state conditions.


Assuntos
Liberação Controlada de Fármacos , Absorção Gástrica/fisiologia , Ibuprofeno/farmacocinética , Período Pós-Prandial/fisiologia , Estômago/fisiologia , Administração Oral , Adulto , Área Sob a Curva , Disponibilidade Biológica , Variação Biológica Individual , Variação Biológica da População/fisiologia , Simulação por Computador , Conjuntos de Dados como Assunto , Feminino , Interações Alimento-Droga/fisiologia , Esvaziamento Gástrico/fisiologia , Voluntários Saudáveis , Humanos , Concentração de Íons de Hidrogênio , Ibuprofeno/administração & dosagem , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Solubilidade , Comprimidos , Adulto Jovem
6.
Mol Pharm ; 14(12): 4295-4304, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-28937221

RESUMO

In vivo drug dissolution in the gastrointestinal (GI) tract is largely unmeasured. The purpose of this clinical study was to evaluate the in vivo drug dissolution and systemic absorption of the BCS class IIa drug ibuprofen under fed and fasted conditions by direct sampling of stomach and small intestinal luminal content. Expanding current knowledge of drug dissolution in vivo will help to establish physiologically relevant in vitro models predictive of drug dissolution. A multilumen GI catheter was orally inserted into the GI tract of healthy human subjects. Subjects received a single oral dose of ibuprofen (800 mg tablet) with 250 mL of water under fasting and fed conditions. The GI catheter facilitated collection of GI fluid from the stomach, duodenum, and jejunum. Ibuprofen concentration in GI fluid supernatant and plasma was determined by LC-MS/MS. A total of 23 subjects completed the study, with 11 subjects returning for an additional study visit (a total of 34 completed study visits). The subjects were primarily white (61%) and male (65%) with an average age of 30 years. The subjects had a median [min, max] weight of 79 [52, 123] kg and body mass index of 25.7 [19.4, 37.7] kg/m2. Ibuprofen plasma levels were higher under fasted conditions and remained detectable for 28 h under both conditions. The AUC0-24 and Cmax were lower in fed subjects vs fasted subjects, and Tmax was delayed in fed subjects vs fasted subjects. Ibuprofen was detected immediately after ingestion in the stomach under fasting and fed conditions until 7 h after dosing. Higher levels of ibuprofen were detected in the small intestine soon after dosing in fasted subjects compared to fed. In contrast to plasma drug concentration, overall gastric concentrations remained higher under fed conditions due to increased gastric pH vs fasting condition. The gastric pH increased to near neutrality after feedingbefore decreasing to acidic levels after 7 h. Induction of the fed state reduced systemic levels but increased gastric levels of ibuprofen, which suggest that slow gastric emptying and transit dominate the effect for plasma drug concentration. The finding of high levels of ibuprofen in stomach and small intestine 7 h post dosing was unexpected. Future work is needed to better understand the role of various GI parameters, such as motility and gastric emptying, on systemic ibuprofen levels in order to improve in vitro predictive models.


Assuntos
Absorção Fisiológica/fisiologia , Liberação Controlada de Fármacos/fisiologia , Trato Gastrointestinal/fisiologia , Ibuprofeno/farmacocinética , Administração Oral , Adulto , Disponibilidade Biológica , Biofarmácia , Jejum/fisiologia , Feminino , Esvaziamento Gástrico/fisiologia , Voluntários Saudáveis , Humanos , Absorção Intestinal/fisiologia , Masculino , Pessoa de Meia-Idade , Período Pós-Prandial , Solubilidade , Comprimidos , Adulto Jovem
7.
Mol Pharm ; 14(2): 345-358, 2017 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-28009518

RESUMO

As an orally administered, locally acting gastrointestinal drug, mesalamine products are designed to achieve high local drug concentration in the gastrointestinal (GI) tract for the treatment of ulcerative colitis. The aim of this study was to directly measure and compare drug dissolution of three mesalamine formulations in human GI tract and to correlate their GI concentration with drug concentration in plasma. Healthy human subjects were orally administered Pentasa, Apriso, or Lialda. GI fluids were aspirated from stomach, duodenum, proximal jejunum, mid jejunum, and distal jejunum regions. Mesalamine (5-ASA) and its primary metabolite acetyl-5-mesalamine (Ac-5-ASA) were measured using LC-MS/MS. GI tract pH was measured from each GI fluid sample, which averaged 1.82, 4.97, 5.67, 6.17, and 6.62 in the stomach, duodenum, proximal jejunum, middle jejunum, and distal jejunum, respectively. For Pentasa, high levels of 5-ASA in solution were observed in the stomach, duodenum, proximal jejunum, mid jejunum, and distal jejunum from 1 to 7 h. Apriso had minimal 5-ASA levels in stomach, low to medium levels of 5-ASA in duodenum and proximal jejunum from 4 to 7 h, and high levels of 5-ASA in distal jejunum from 3 to 7 h. In contrast, Lialda had minimal 5-ASA levels from stomach and early small intestine. A composite appearance rate (CAR) was calculated from the deconvolution of individual plasma concentration to reflect drug release, dissolution, transit, and absorption in the GI tract. Individuals dosed with Pentasa had high levels of CAR from 1 to 10 h; individuals dosed with Apriso had low levels of CAR from 1 to 4 h and high levels of CAR from 5 to 10 h; Lialda showed minimal levels of CAR from 0 to 5 h, then increased to medium levels from 5 to 12 h, and then decreased to further lower levels after 12 h. In the colon region, Pentasa and Apriso showed similar levels of accumulated 5-ASA excreted in the feces, while Lialda showed slightly higher 5-ASA accumulation in feces. However, all three formulations showed similar levels of metabolite Ac-5-ASA in the feces. These results provide direct measurement of drug dissolution in the GI tract, which can serve as a basis for investigation of bioequivalence for locally acting drug products.


Assuntos
Liberação Controlada de Fármacos/fisiologia , Trato Gastrointestinal/metabolismo , Mesalamina/metabolismo , Administração Oral , Adolescente , Adulto , Química Farmacêutica/métodos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Solubilidade , Adulto Jovem
8.
Mol Pharm ; 14(12): 4281-4294, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-28737409

RESUMO

In this study, we determined the pH and buffer capacity of human gastrointestinal (GI) fluids (aspirated from the stomach, duodenum, proximal jejunum, and mid/distal jejunum) as a function of time, from 37 healthy subjects after oral administration of an 800 mg immediate-release tablet of ibuprofen (reference listed drug; RLD) under typical prescribed bioequivalence (BE) study protocol conditions in both fasted and fed states (simulated by ingestion of a liquid meal). Simultaneously, motility was continuously monitored using water-perfused manometry. The time to appearance of phase III contractions (i.e., housekeeper wave) was monitored following administration of the ibuprofen tablet. Our results clearly demonstrated the dynamic change in pH as a function of time and, most significantly, the extremely low buffer capacity along the GI tract. The buffer capacity on average was 2.26 µmol/mL/ΔpH in fasted state (range: 0.26 and 6.32 µmol/mL/ΔpH) and 2.66 µmol/mL/ΔpH in fed state (range: 0.78 and 5.98 µmol/mL/ΔpH) throughout the entire upper GI tract (stomach, duodenum, and proximal and mid/distal jejunum). The implication of this very low buffer capacity of the human GI tract is profound for the oral delivery of both acidic and basic active pharmaceutical ingredients (APIs). An in vivo predictive dissolution method would require not only a bicarbonate buffer but also, more significantly, a low buffer capacity of dissolution media to reflect in vivo dissolution conditions.


Assuntos
Líquidos Corporais/química , Motilidade Gastrointestinal/fisiologia , Trato Gastrointestinal/fisiologia , Ibuprofeno/farmacocinética , Absorção Intestinal/fisiologia , Absorção Fisiológica , Administração Oral , Adulto , Líquidos Corporais/fisiologia , Soluções Tampão , Liberação Controlada de Fármacos , Voluntários Saudáveis , Humanos , Concentração de Íons de Hidrogênio , Mucosa Intestinal/fisiologia , Manometria , Pessoa de Meia-Idade , Solubilidade , Comprimidos , Equivalência Terapêutica , Fatores de Tempo , Adulto Jovem
9.
Infect Immun ; 83(3): 934-41, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25534943

RESUMO

Clostridium difficile infection (CDI) following antibiotic therapy is a major public health threat. While antibiotic disruption of the indigenous microbiota underlies the majority of cases of CDI, the early dynamics of infection in the disturbed intestinal ecosystem are poorly characterized. This study defines the dynamics of infection with C. difficile strain VPI 10463 throughout the gastrointestinal (GI) tract using a murine model of infection. After inducing susceptibility to C. difficile colonization via antibiotic administration, we followed the dynamics of spore germination, colonization, sporulation, toxin activity, and disease progression throughout the GI tract. C. difficile spores were able to germinate within 6 h postchallenge, resulting in the establishment of vegetative bacteria in the distal GI tract. Spores and cytotoxin activity were detected by 24 h postchallenge, and histopathologic colitis developed by 30 h. Within 36 h, all infected mice succumbed to infection. We correlated the establishment of infection with changes in the microbiota and bile acid profile of the small and large intestines. Antibiotic administration resulted in significant changes to the microbiota in the small and large intestines, as well as a significant shift in the abundance of primary and secondary bile acids. Ex vivo analysis suggested the small intestine as the site of spore germination. This study provides an integrated understanding of the timing and location of the events surrounding C. difficile colonization and identifies potential targets for the development of new therapeutic strategies.


Assuntos
Clostridioides difficile/patogenicidade , Infecções por Clostridium/patologia , Colite/patologia , Trato Gastrointestinal/patologia , Animais , Antibacterianos/efeitos adversos , Ácidos e Sais Biliares/química , Clostridioides difficile/crescimento & desenvolvimento , Clostridioides difficile/metabolismo , Infecções por Clostridium/etiologia , Infecções por Clostridium/microbiologia , Infecções por Clostridium/mortalidade , Colite/etiologia , Colite/microbiologia , Colite/mortalidade , Progressão da Doença , Enterotoxinas/biossíntese , Enterotoxinas/metabolismo , Fezes/microbiologia , Feminino , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microbiota/efeitos dos fármacos , Esporos Bacterianos/crescimento & desenvolvimento , Esporos Bacterianos/metabolismo , Esporos Bacterianos/patogenicidade , Análise de Sobrevida , Fatores de Tempo
10.
Curr Opin Gastroenterol ; 29(6): 628-32, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24100717

RESUMO

PURPOSE OF REVIEW: The use of faecal microbiota transplantation (FMT) as treatment for recurrent Clostridium difficile infection (CDI) has increased rapidly over the past few years. In this review, we highlight clinical studies of FMT for treatment of recurrent CDI and discuss the safety, standardization and future of this treatment option. The major risk factor for CDI is prior antibiotic use, which results in an altered state of the gut microbiota characterized by decreased microbial diversity. This altered gut microbiota increases the patient's susceptibility to CDI. In patients with recurrent CDI, the microbiota remains in a state with decreased diversity, and FMT from a healthy individual restores the gut microbiota and subsequently colonization resistance against the pathogen. RECENT FINDINGS: Recent studies have shown the success rate for FMT as treatment for recurrent CDI being greater than 90%. Standardized, frozen preparations of faeces can be used, which increases the availability of faeces for FMT and decreases the cost of screening individual donors. In addition, there have been recent advances in identifying a defined microbial community isolated from faeces that can restore colonization resistance against C. difficile. SUMMARY: The use of FMT is a successful treatment for recurrent CDI when primary treatment options have failed. However, more work needs to define potential long-term consequences of this treatment and understand how specific members of the gut microbiota can restore colonization resistance against C. difficile.


Assuntos
Terapia Biológica/métodos , Enterocolite Pseudomembranosa/terapia , Fezes/microbiologia , Microbiota , Terapia Biológica/efeitos adversos , Terapia Biológica/normas , Enterocolite Pseudomembranosa/microbiologia , Humanos , Intestinos/microbiologia , Ensaios Clínicos Controlados Aleatórios como Assunto , Recidiva
11.
Infect Immun ; 80(11): 3786-94, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22890996

RESUMO

The indigenous microbial community of the gastrointestinal (GI) tract determines susceptibility to Clostridium difficile colonization and disease. Previous studies have demonstrated that antibiotic-treated mice challenged with C. difficile either developed rapidly lethal C. difficile infection or were stably colonized with mild disease. The GI microbial community of animals with mild disease was dominated by members of the bacterial family Lachnospiraceae, while the gut community in moribund animals had a predominance of Escherichia coli. We investigated the roles of murine Lachnospiraceae and E. coli strains in colonization resistance against C. difficile in germfree mice. Murine Lachnospiraceae and E. coli isolates were cultured from wild-type mice. The ability of each of these isolates to interfere with C. difficile colonization was tested by precolonizing germfree mice with these bacteria 4 days prior to experimental C. difficile challenge. Mice precolonized with a murine Lachnospiraceae isolate, but not those colonized with E. coli, had significantly decreased C. difficile colonization, lower intestinal cytotoxin levels and exhibited less severe clinical signs and colonic histopathology. Infection of germfree mice or mice precolonized with E. coli with C. difficile strain VPI 10463 was uniformly fatal by 48 h, but only 20% mortality was seen at 2 days in mice precolonized with the Lachnospiraceae isolate prior to challenge with VPI 10463. These findings confirm that a single component of the GI microbiota, a murine Lachnospiraceae isolate, could partially restore colonization resistance against C. difficile. Further study of the members within the Lachnospiraceae family could lead to a better understanding of mechanisms of colonization resistance against C. difficile and novel therapeutic approaches for the treatment and prevention of C. difficile infection.


Assuntos
Clostridioides difficile/patogenicidade , Trato Gastrointestinal/microbiologia , Animais , Clostridioides difficile/genética , Contagem de Colônia Microbiana , Escherichia coli/genética , Escherichia coli/isolamento & purificação , Vida Livre de Germes , Bacilos Gram-Positivos Formadores de Endosporo/genética , Bacilos Gram-Positivos Formadores de Endosporo/isolamento & purificação , Metagenoma , Camundongos , Camundongos Endogâmicos C57BL , Filogenia , RNA Ribossômico 16S
12.
Microbiology (Reading) ; 156(Pt 3): 950-959, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19959576

RESUMO

Phosphoribosylamine (PRA) is the first intermediate in the common purine/thiamine biosynthetic pathway and is primarily synthesized by the product of the purF gene, glutamine phosphoribosylpyrophosphate (PRPP) amidotransferase (E.C. 2.4.2.14). Past genetic and biochemical studies have shown that multiple mechanisms for the synthesis of PRA independent of PurF are present in Salmonella enterica. Here, we describe mutant alleles of the essential prsA gene, which encodes PRPP synthetase (E.C. 2.7.6.1), that allow PurF-independent thiamine synthesis. The mutant alleles resulted in reduced PrsA activity in extracts, caused nutritional requirements indicative of PRPP limitation and allowed non-enzymic formation of PRA due to a build-up of ribose 5-phosphate (R5P). These results emphasize the balance that must be reached between pathways competing for the same substrate to maintain robustness of the metabolic network.


Assuntos
Proteínas de Bactérias/metabolismo , Lipoproteínas/metabolismo , Proteínas de Membrana/metabolismo , Ribosemonofosfatos/metabolismo , Salmonella enterica/enzimologia , Tiamina/biossíntese , Transaminases/metabolismo , Alelos , Proteínas de Bactérias/genética , Meios de Cultura , Lipoproteínas/genética , Proteínas de Membrana/genética , Redes e Vias Metabólicas , Mutação , Salmonella enterica/genética , Salmonella enterica/crescimento & desenvolvimento
13.
AAPS J ; 22(1): 3, 2019 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-31712917

RESUMO

Multiple approaches such as mathematical deconvolution and mechanistic oral absorption models have been used to predict in vivo drug dissolution in the gastrointestinal (GI) tract. However, these approaches are often validated by plasma pharmacokinetic profiles, but not by in vivo drug dissolution due to the limited data available regarding the local GI environment. It is also challenging to predict and validate in vivo dissolution in different regions of the GI tract (stomach, duodenum, jejunum, and ileum). In this study, the dynamic fluid compartment absorption and transport (DFCAT) model was used to predict the in vivo dissolution profiles of ibuprofen, which was administered as an 800-mg immediate-release tablet to healthy subjects, in different regions of the GI tract. The prediction was validated with concentration time-courses of ibuprofen (BCS class 2a) in different regions of the GI tract that we have obtained over the past few years. The computational model predicted that the dissolution of ibuprofen was minimal in the stomach (2%), slightly more in the duodenum (6.3%), and primarily dissolved in the jejunum (63%) and the ileum (25%). The detailed model prediction of drug dissolution in different regions of GI can provide a quantitative reference of in vivo dissolution that may provide valuable insight in developing in vitro tests for drug product optimization and quality.


Assuntos
Anti-Inflamatórios não Esteroides/farmacocinética , Liberação Controlada de Fármacos , Ibuprofeno/farmacocinética , Absorção Intestinal , Modelos Teóricos , Administração Oral , Anti-Inflamatórios não Esteroides/administração & dosagem , Trato Gastrointestinal , Humanos , Ibuprofeno/administração & dosagem , Estudo de Prova de Conceito
14.
mSphere ; 4(2)2019 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-30867328

RESUMO

Although the microbiota in the proximal gastrointestinal (GI) tract have been implicated in health and disease, much about these microbes remains understudied compared to those in the distal GI tract. This study characterized the microbiota across multiple proximal GI sites over time in healthy individuals. As part of a study of the pharmacokinetics of oral mesalamine administration, healthy, fasted volunteers (n = 8; 10 observation periods total) were orally intubated with a four-lumen catheter with multiple aspiration ports. Samples were taken from stomach, duodenal, and multiple jejunal sites, sampling hourly (≤7 h) to measure mesalamine (administered at t = 0), pH, and 16S rRNA gene-based composition. We observed a predominance of Firmicutes across proximal GI sites, with significant variation compared to stool. The microbiota was more similar within individuals over time than between subjects, with the fecal microbiota being unique from that of the small intestine. The stomach and duodenal microbiota displayed highest intraindividual variability compared to jejunal sites, which were more stable across time. We observed significant correlations in the duodenal microbial composition with changes in pH; linear mixed models identified positive correlations with multiple Streptococcus operational taxonomic units (OTUs) and negative correlations with multiple Prevotella and Pasteurellaceae OTUs. Few OTUs correlated with mesalamine concentration. The stomach and duodenal microbiota exhibited greater compositional dynamics than the jejunum. Short-term fluctuations in the duodenal microbiota were correlated with pH. Given the unique characteristics and dynamics of the proximal GI tract microbiota, it is important to consider these local environments in health and disease states.IMPORTANCE The gut microbiota are linked to a variety of gastrointestinal diseases, including inflammatory bowel disease. Despite this importance, microbiota dynamics in the upper gastrointestinal tract are understudied. Our article seeks to understand what factors impact microbiota dynamics in the healthy human upper gut. We found that the upper gastrointestinal tract contains consistently prevalent bacterial OTUs that dominate the overall community. Microbiota variability is highest in the stomach and duodenum and correlates with pH.


Assuntos
Bactérias/classificação , Jejum , Microbioma Gastrointestinal , Intestino Delgado/microbiologia , Estômago/microbiologia , Administração Oral , Adolescente , Adulto , Bactérias/isolamento & purificação , Fezes/microbiologia , Feminino , Firmicutes/classificação , Firmicutes/isolamento & purificação , Voluntários Saudáveis , Humanos , Concentração de Íons de Hidrogênio , Intubação Gastrointestinal , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Pasteurellaceae/classificação , Pasteurellaceae/isolamento & purificação , RNA Ribossômico 16S/genética , Análise Espaço-Temporal , Adulto Jovem
15.
Eur J Pharm Biopharm ; 129: 162-174, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29857136

RESUMO

The goal of this study was to create a mass transport model (MTM) model for gastric emptying and upper gastrointestinal (GI) appearance that can capture the in vivo concentration-time profiles of the nonabsorbable drug phenol red in solution in the stomach and upper small intestine by direct luminal measurement while simultaneously recording the contractile activity (motility) via manometry. We advanced from a one-compartmental design of the stomach to a much more appropriate, multi-compartmental 'mixing tank' gastric model that reflects drug distribution along the different regions of the stomach as a consequence of randomly dosing relative to the different contractile phases of the migrating motor complex (MMC). To capture the intraluminal phenol red concentrations in the different segments of the GI tract both in fasted and fed state conditions, it was essential to include a bypass flow compartment ('magenstrasse') to facilitate the transport of the phenol red solution directly to the duodenum (fasted state) or antrum (fed state). The fasted and fed state models were validated with external reference data from an independent aspiration study using another nonabsorbable marker (paromomycin). These results will be essential for the development and optimization of computational programs for GI simulation and absorption prediction, providing a realistic gastric physiologically-based pharmacokinetic (PBPK) model based on direct measurement of gastric concentrations of the drug in the stomach.


Assuntos
Esvaziamento Gástrico/efeitos dos fármacos , Absorção Intestinal , Intestino Delgado/efeitos dos fármacos , Modelos Biológicos , Estômago/fisiologia , Administração Oral , Adulto , Jejum , Feminino , Voluntários Saudáveis , Humanos , Intestino Delgado/fisiologia , Masculino , Pessoa de Meia-Idade , Paromomicina/farmacologia , Fenolsulfonaftaleína/farmacologia , Período Pós-Prandial , Solubilidade , Estômago/efeitos dos fármacos , Adulto Jovem
16.
Cell Host Microbe ; 24(3): 364-378.e6, 2018 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-30212649

RESUMO

In addition to high-fat diet (HFD) and inactivity, inflammation and microbiota composition contribute to obesity. Inhibitory immune receptors, such as NLRP12, dampen inflammation and are important for resolving inflammation, but their role in obesity is unknown. We show that obesity in humans correlates with reduced expression of adipose tissue NLRP12. Similarly, Nlrp12-/- mice show increased weight gain, adipose deposition, blood glucose, NF-κB/MAPK activation, and M1-macrophage polarization. Additionally, NLRP12 is required to mitigate HFD-induced inflammasome activation. Co-housing with wild-type animals, antibiotic treatment, or germ-free condition was sufficient to restrain inflammation, obesity, and insulin tolerance in Nlrp12-/- mice, implicating the microbiota. HFD-fed Nlrp12-/- mice display dysbiosis marked by increased obesity-associated Erysipelotrichaceae, but reduced Lachnospiraceae family and the associated enzymes required for short-chain fatty acid (SCFA) synthesis. Lachnospiraceae or SCFA administration attenuates obesity, inflammation, and dysbiosis. These findings reveal that Nlrp12 reduces HFD-induced obesity by maintaining beneficial microbiota.


Assuntos
Microbioma Gastrointestinal , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Obesidade/imunologia , Obesidade/microbiologia , Tecido Adiposo/imunologia , Adulto , Idoso , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Bactérias/metabolismo , Feminino , Homeostase , Humanos , Imunidade Inata , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Obesidade/genética , Obesidade/metabolismo
17.
Nat Microbiol ; 2: 16267, 2017 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-28112760

RESUMO

Mesalamine serves as the gold standard in treating ulcerative colitis. However, its precise mechanism(s) of action remains unclear. Here, we show that mesalamine treatment rapidly decreases polyphosphate levels in diverse bacteria, including members of the human gut microbiome. This decrease sensitizes bacteria towards oxidative stress, reduces colonization and attenuates persister cell and biofilm formation, suggesting that mesalamine aids in diminishing the capacity of bacteria to persist within chronically inflamed environments.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Negativas/fisiologia , Mesalamina/farmacologia , Polifosfatos/metabolismo , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/uso terapêutico , Biofilmes/efeitos dos fármacos , Ceco/microbiologia , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/microbiologia , Escherichia coli/efeitos dos fármacos , Fezes/microbiologia , Bactérias Gram-Negativas/genética , Humanos , Mesalamina/administração & dosagem , Mesalamina/uso terapêutico , Camundongos , Estresse Oxidativo/efeitos dos fármacos
18.
Nat Commun ; 5: 3114, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24445449

RESUMO

Antibiotics can have significant and long-lasting effects on the gastrointestinal tract microbiota, reducing colonization resistance against pathogens including Clostridium difficile. Here we show that antibiotic treatment induces substantial changes in the gut microbial community and in the metabolome of mice susceptible to C. difficile infection. Levels of secondary bile acids, glucose, free fatty acids and dipeptides decrease, whereas those of primary bile acids and sugar alcohols increase, reflecting the modified metabolic activity of the altered gut microbiome. In vitro and ex vivo analyses demonstrate that C. difficile can exploit specific metabolites that become more abundant in the mouse gut after antibiotics, including the primary bile acid taurocholate for germination, and carbon sources such as mannitol, fructose, sorbitol, raffinose and stachyose for growth. Our results indicate that antibiotic-mediated alteration of the gut microbiome converts the global metabolic profile to one that favours C. difficile germination and growth.


Assuntos
Antibacterianos/farmacologia , Clostridioides difficile/fisiologia , Infecções por Clostridium/metabolismo , Infecções por Clostridium/microbiologia , Trato Gastrointestinal/microbiologia , Metaboloma/efeitos dos fármacos , Microbiota/efeitos dos fármacos , Animais , Clostridioides difficile/efeitos dos fármacos , Clostridioides difficile/crescimento & desenvolvimento , Suscetibilidade a Doenças/metabolismo , Suscetibilidade a Doenças/microbiologia , Feminino , Masculino , Metabolômica , Camundongos Endogâmicos C57BL
19.
PLoS One ; 7(10): e48207, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23133571

RESUMO

Phosphoribosylamine (PRA) is an intermediate in the biosynthetic pathway that is common to thiamine and purines. Glutamine phosphoribosyl pyrophosphate (PRPP) amidotransferase is the product of the purF gene in Salmonella enterica and catalyzes the synthesis of PRA from PRPP and glutamine. Strains lacking PurF require exogenous addition of purines for growth. However, under some growth conditions or with specific secondary mutations these strains grow in the absence of exogenous thiamine. Mutant alleles of hisA, which encodes 1-(5-phosphoribosyl)-5-[(5-phosphoribosylamino) methylideneamino] imidazole-4-carboxamide (ProFAR) isomerase, allowed PurF-independent PRA formation. The alleles of hisA that suppressed the requirement for exogenous thiamine resulted in proteins with reduced enzymatic activity. Data presented here showed that decreased activity of HisA altered metabolite pools and allowed PRA formation from ProFAR. Possible mechanisms of this conversion were proposed. The results herein emphasize the plasticity of the metabolic network and specifically highlight the potential for chemical syntheses to contribute to network robustness.


Assuntos
Amidofosforribosiltransferase/genética , Histidina/metabolismo , Salmonella enterica/metabolismo , Alelos , Amidofosforribosiltransferase/metabolismo , Cromatografia Líquida de Alta Pressão/métodos , DNA/metabolismo , Histidina/química , Redes e Vias Metabólicas/fisiologia , Modelos Químicos , Modelos Genéticos , Mutação , Óperon , Purinas/metabolismo , Tiamina/metabolismo
20.
Trends Microbiol ; 18(6): 240-7, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20382023

RESUMO

The emergence of systems biology has re-emphasized the advantages of understanding biological processes with a global perspective. One biological process amenable to global approaches is microbial metabolism. This review describes a model system that contributes to the goals of systems biology by experimentally defining metabolic integration found in a bacterial cell and thus providing data needed for implementation and interpretation of systems approaches. We have taken a largely unbiased in vivo approach centered on thiamine biosynthesis to identify new metabolic components and connections, and to explore uncharacterized paradigms of the integration between them. This article summarizes recent results from this approach that include the identification of the function of unknown genes, connections between cofactors biosynthesis and thiamine biosynthesis, and how metabolites from one biosynthetic pathway can be used in thiamine biosynthesis.


Assuntos
Bactérias/metabolismo , Salmonella enterica/metabolismo , Tiamina/biossíntese , Bactérias/genética , Vias Biossintéticas , Genes Bacterianos , Genes Reguladores , Modelos Biológicos , Ribose-Fosfato Pirofosfoquinase/metabolismo , Salmonella enterica/genética , Biologia de Sistemas/métodos , Tiamina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA