Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Immunity ; 43(3): 527-40, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26362264

RESUMO

The interrelationship between IgAs and microbiota diversity is still unclear. Here we show that BALB/c mice had higher abundance and diversity of IgAs than C57BL/6 mice and that this correlated with increased microbiota diversity. We show that polyreactive IgAs mediated the entrance of non-invasive bacteria to Peyer's patches, independently of CX3CR1(+) phagocytes. This allowed the induction of bacteria-specific IgA and the establishment of a positive feedback loop of IgA production. Cohousing of mice or fecal transplantation had little or no influence on IgA production and had only partial impact on microbiota composition. Germ-free BALB/c, but not C57BL/6, mice already had polyreactive IgAs that influenced microbiota diversity and selection after colonization. Together, these data suggest that genetic predisposition to produce polyreactive IgAs has a strong impact on the generation of antigen-specific IgAs and the selection and maintenance of microbiota diversity.


Assuntos
Antígenos de Bactérias/imunologia , Variação Genética/imunologia , Imunoglobulina A/imunologia , Microbiota/imunologia , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/imunologia , DNA Bacteriano/química , DNA Bacteriano/genética , Fezes/microbiologia , Citometria de Fluxo , Interações Hospedeiro-Patógeno/imunologia , Imunização , Imunoglobulina A/sangue , Imunoglobulina A/metabolismo , Metagenômica/métodos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microbiota/genética , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/metabolismo , Nódulos Linfáticos Agregados/microbiologia , Filogenia , RNA Ribossômico 16S/genética , Salmonella typhimurium/genética , Salmonella typhimurium/imunologia , Salmonella typhimurium/fisiologia , Especificidade da Espécie
2.
PLoS Biol ; 19(1): e3001070, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33481771

RESUMO

Microbial conversion of dietary or drug substrates into small bioactive molecules represents a regulatory mechanism by which the gut microbiota alters intestinal physiology. Here, we show that a wide variety of gut bacteria can metabolize the dietary supplement and antidepressant 5-hydroxytryptophan (5-HTP) to 5-hydroxyindole (5-HI) via the tryptophanase (TnaA) enzyme. Oral administration of 5-HTP results in detection of 5-HI in fecal samples of healthy volunteers with interindividual variation. The production of 5-HI is inhibited upon pH reduction in in vitro studies. When administered orally in rats, 5-HI significantly accelerates the total gut transit time (TGTT). Deciphering the underlying mechanisms of action reveals that 5-HI accelerates gut contractility via activation of L-type calcium channels located on the colonic smooth muscle cells. Moreover, 5-HI stimulation of a cell line model of intestinal enterochromaffin cells results in significant increase in serotonin production. Together, our findings support a role for bacterial metabolism in altering gut motility and lay the foundation for microbiota-targeted interventions.


Assuntos
Bactérias/metabolismo , Canais de Cálcio Tipo L/efeitos dos fármacos , Motilidade Gastrointestinal/efeitos dos fármacos , Indóis/metabolismo , Indóis/farmacologia , 5-Hidroxitriptofano/metabolismo , Adulto , Animais , Canais de Cálcio Tipo L/metabolismo , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Motilidade Gastrointestinal/fisiologia , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Masculino , Ratos , Adulto Jovem
3.
BMC Biol ; 18(1): 137, 2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-33076930

RESUMO

BACKGROUND: Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by both motor and non-motor symptoms. Gastrointestinal tract dysfunction is one of the non-motor features, where constipation is reported as the most common gastrointestinal symptom. Aromatic bacterial metabolites are attracting considerable attention due to their impact on gut homeostasis and host's physiology. In particular, Clostridium sporogenes is a key contributor to the production of these bioactive metabolites in the human gut. RESULTS: Here, we show that C. sporogenes deaminates levodopa, the main treatment in Parkinson's disease, and identify the aromatic aminotransferase responsible for the initiation of the deamination pathway. The deaminated metabolite from levodopa, 3-(3,4-dihydroxyphenyl)propionic acid, elicits an inhibitory effect on ileal motility in an ex vivo model. We detected 3-(3,4-dihydroxyphenyl)propionic acid in fecal samples of Parkinson's disease patients on levodopa medication and found that this metabolite is actively produced by the gut microbiota in those stool samples. CONCLUSIONS: Levodopa is deaminated by the gut bacterium C. sporogenes producing a metabolite that inhibits ileal motility ex vivo. Overall, this study underpins the importance of the metabolic pathways of the gut microbiome involved in drug metabolism not only to preserve drug effectiveness, but also to avoid potential side effects of bacterial breakdown products of the unabsorbed residue of medication.


Assuntos
Antiparkinsonianos/metabolismo , Clostridium/metabolismo , Motilidade Gastrointestinal , Levodopa/metabolismo , Transaminases/metabolismo , Animais , Antiparkinsonianos/química , Clostridium/enzimologia , Desaminação , Microbioma Gastrointestinal , Levodopa/química , Masculino , Camundongos/microbiologia , Camundongos Endogâmicos C57BL , Doença de Parkinson/tratamento farmacológico
4.
Cell Mol Neurobiol ; 40(2): 191-201, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31836967

RESUMO

Trace amines and their primary receptor, Trace Amine-Associated Receptor-1 (TAAR1) are widely studied for their involvement in the pathogenesis of neuropsychiatric disorders despite being found in the gastrointestinal tract at physiological levels. With the emergence of the "brain-gut-microbiome axis," we take the opportunity to review what is known about trace amines in the brain, the defined sources of trace amines in the gut, and emerging understandings on the levels of trace amines in various gastrointestinal disorders. Similarly, we discuss localization of TAAR1 expression in the gut, novel findings that TAAR1 may be implicated in inflammatory bowel diseases, and the reported comorbidities of neuropsychiatric disorders and gastrointestinal disorders. With the emergence of TAAR1 specific compounds as next-generation therapeutics for schizophrenia (Roche) and Parkinson's related psychoses (Sunovion), we hypothesize a therapeutic benefit of these compounds in clinical trials in the brain-gut-microbiome axis, as well as a potential for thoughtful manipulation of the brain-gut-microbiome axis to modulate symptoms of neuropsychiatric disease.


Assuntos
Encéfalo/metabolismo , Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/metabolismo , Transtornos Mentais/metabolismo , Receptores Acoplados a Proteínas G/biossíntese , Animais , Trato Gastrointestinal/microbiologia , Humanos , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/psicologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Transtornos Mentais/psicologia
5.
J Sleep Res ; 29(6): e12920, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-31515894

RESUMO

Not getting enough sleep is a common problem in our society and contributes to numerous health problems, including high blood pressure, diabetes and obesity. Related to these observations, a wealth of studies has underscored the negative impact of both acute and chronic sleep deprivation on cognitive function. More recently it has become apparent that the gut microbiota composition can be rapidly altered, modulates brain function and is affected by the aforementioned health problems. As such, changes in the microbiota composition may contribute to the behavioural and physiological phenotypes associated with sleep deprivation. It is unclear, however, whether a brief period of sleep deprivation can also negatively impact the gut microbiota. Here, we examined the impact of 5 hr of sleep deprivation on gut microbiota composition of male C57Bl6/J mice. Despite the fact that the overall microbial composition did not change between the control- and sleep-deprived groups, the relative abundance of the Clostridiaceae and Lachnospiraceae were slightly altered in sleep-deprived animals compared to controls. Together, these data suggest that depriving mice of sleep for 5 hr leads to subtle changes in the gut microbiota composition.


Assuntos
Microbioma Gastrointestinal/fisiologia , Privação do Sono/complicações , Animais , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL
6.
Adv Exp Med Biol ; 874: 301-36, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26589226

RESUMO

The gut microbiome plays a crucial role in host physiology. Disruption of its community structure and function can have wide-ranging effects making it critical to understand exactly how the interactive dialogue between the host and its microbiota is regulated to maintain homeostasis. An array of multidirectional signalling molecules is clearly involved in the host-microbiome communication. This interactive signalling not only impacts the gastrointestinal tract, where the majority of microbiota resides, but also extends to affect other host systems including the brain and liver as well as the microbiome itself. Understanding the mechanistic principles of this inter-kingdom signalling is fundamental to unravelling how our supraorganism function to maintain wellbeing, subsequently opening up new avenues for microbiome manipulation to favour desirable mental health outcome.


Assuntos
Encéfalo/fisiologia , Microbioma Gastrointestinal/fisiologia , Interações Hospedeiro-Patógeno , Animais , Epigênese Genética , Humanos , Transdução de Sinais
7.
Gut ; 64(6): 884-93, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25056659

RESUMO

OBJECTIVE: Repetitive interaction with microbial stimuli renders epithelial cells (ECs) hyporesponsive to microbial stimulation. Previously, we have reported that buccal ECs from a subset of paediatric patients with Crohn's disease are not hyporesponsive and spontaneously released chemokines. We now aimed to identify kinetics and mechanisms of acquisition of hyporesponsiveness to microbial stimulation using primary human buccal epithelium. DESIGN: Buccal ECs collected directly after birth and in later stages of life were investigated. Chemokine release and regulatory signalling pathways were studied using primary buccal ECs and the buccal EC line TR146. Findings were extended to the intestinal mucosa using murine model systems. RESULTS: Directly after birth, primary human buccal ECs spontaneously produced the chemokine CXCL-8 and were responsive to microbial stimuli. Within the first weeks of life, these ECs attained hyporesponsiveness, associated with inactivation of the NF-κB pathway and upregulation of the novel NF-κB inhibitor SLPI but no other known NF-κB inhibitors. SLPI protein was abundant in the cytoplasm and the nucleus of hyporesponsive buccal ECs. Knock-down of SLPI in TR146-buccal ECs induced loss of hyporesponsiveness with increased NF-κB activation and subsequent chemokine release. This regulatory mechanism extended to the intestine, as colonisation of germfree mice elicited SLPI expression in small intestine and colon. Moreover, SLPI-deficient mice had increased chemokine expression in small intestinal and colonic ECs. CONCLUSIONS: We identify SLPI as a new player in acquisition of microbial hyporesponsiveness by buccal and intestinal epithelium in the first weeks after microbial colonisation.


Assuntos
Envelhecimento/imunologia , Epitélio/imunologia , Epitélio/microbiologia , Mucosa Bucal/citologia , Mucosa Bucal/microbiologia , Inibidor Secretado de Peptidases Leucocitárias/metabolismo , Adulto , Animais , Células Cultivadas , Quimiocina CXCL2/metabolismo , Regulação para Baixo , Epitélio/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Tolerância Imunológica , Lactente , Recém-Nascido , Interleucina-8/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/microbiologia , Camundongos , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Peptidoglicano/farmacologia
8.
Bioessays ; 35(10): 913-23, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23946088

RESUMO

In this review, we provide an overview of the dynamic changes within the microbiota and its metabolites that are implicated in establishing and maintaining gastrointestinal homeostasis during various stages of microbial colonization. The gradual conversion of the gut microbiota toward a mutualistic microbial community involves replacement of pioneer gut colonizers with bacterial taxa that are characteristic for the adult gut. An important microbial signature of homeostasis in the adult gut is the prevalence and activity of a diverse spectrum of bacterial species that produce beneficial metabolites through metabolic interactions between microbial groups. Deciphering these microbial signatures and their metabolites that govern short and long-term equilibrium, as well as imbalances in host-microbial relationships, may provide novel diagnostic tools and/or therapeutic targets for specific disorders associated with intestinal dysbiosis and loss of homeostasis.


Assuntos
Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Microbiota , Animais , Butiratos/metabolismo , Clostridium/classificação , Clostridium/isolamento & purificação , Clostridium/metabolismo , Homeostase , Interações Hospedeiro-Patógeno , Humanos , Metaboloma/fisiologia , Simbiose
9.
Gut ; 62(9): 1306-14, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22722618

RESUMO

OBJECTIVE: Proper interactions between the intestinal mucosa, gut microbiota and nutrient flow are required to establish homoeostasis of the host. Since the proximal part of the small intestine is the first region where these interactions occur, and since most of the nutrient absorption occurs in the jejunum, it is important to understand the dynamics of metabolic responses of the mucosa in this intestinal region. DESIGN: Germ-free mice aged 8-10 weeks were conventionalised with faecal microbiota, and responses of the jejunal mucosa to bacterial colonisation were followed over a 30-day time course. Combined transcriptome, histology, (1)H NMR metabonomics and microbiota phylogenetic profiling analyses were used. RESULTS: The jejunal mucosa showed a two-phase response to the colonising microbiota. The acute-phase response, which had already started 1 day after conventionalisation, involved repression of the cell cycle and parts of the basal metabolism. The secondary-phase response, which was consolidated during conventionalisation (days 4-30), was characterised by a metabolic shift from an oxidative energy supply to anabolic metabolism, as inferred from the tissue transcriptome and metabonome changes. Detailed transcriptome analysis identified tissue transcriptional signatures for the dynamic control of the metabolic reorientation in the jejunum. The molecular components identified in the response signatures have known roles in human metabolic disorders, including insulin sensitivity and type 2 diabetes mellitus. CONCLUSION: This study elucidates the dynamic jejunal response to the microbiota and supports a prominent role for the jejunum in metabolic control, including glucose and energy homoeostasis. The molecular signatures of this process may help to find risk markers in the declining insulin sensitivity seen in human type 2 diabetes mellitus, for instance.


Assuntos
Bactérias/metabolismo , Mucosa Intestinal , Jejuno , Microbiota/fisiologia , Animais , Metabolismo Energético , Fezes/microbiologia , Homeostase , Interações Hospedeiro-Patógeno , Humanos , Absorção Intestinal/fisiologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Jejuno/metabolismo , Jejuno/microbiologia , Jejuno/patologia , Jejuno/fisiopatologia , Metabolômica , Camundongos , Modelos Animais , Filogenia , Fatores de Tempo , Transcriptoma
10.
Curr Opin Gastroenterol ; 29(6): 621-7, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24100720

RESUMO

PURPOSE OF REVIEW: The dynamic interplay of the intestinal microbiota and host has been the focus of many studies because of its impact on the health status in human life. Recent reports on the time-resolved immune and metabolic interactions between the host and microbiota, as well as the molecular signatures that mark this communication during the process of establishing a host-microbial relationship, are addressed here. RECENT FINDINGS: During microbial colonization, the progressive impact of de-novo introduction of the gut microbiota on the host's physiology is tightly controlled by highly intertwined regulatory networks and achieves an efficient and balanced interplay between the host and its developing microbial community. Recent findings from germ-free mouse models have unravelled core transcriptional, metabolic and microbial signatures, which are proposed to orchestrate the molecular responses during the establishment of the multifaceted state of intestinal host-microbe homeostasis. SUMMARY: Exploring the time-resolved dynamics of the host responses to the newly colonizing gut microbiota provides mechanistically critical understanding of the sequential host-microbe response cascades that lead to a homeostatic relationship. Thereby, these approaches can provide novel diagnostic tools and therapeutic targets, or either of the two, in humans for specific disorders associated with intestinal dysbiosis and loss of homeostasis.


Assuntos
Homeostase/fisiologia , Intestinos/microbiologia , Microbiota/fisiologia , Transcriptoma/fisiologia , Animais , Homeostase/genética , Humanos , Intestinos/fisiologia , Microbiota/genética , Modelos Animais , Simbiose/genética , Simbiose/fisiologia
11.
mSystems ; 8(5): e0033223, 2023 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-37668401

RESUMO

IMPORTANCE: The development of probiotic therapies targeted at the small intestinal microbiota represents a significant advancement in the field of probiotic interventions. This region poses unique opportunities due to its low number of gut microbiota, along with the presence of heightened immune and metabolic host responses. However, progress in this area has been hindered by a lack of detailed understanding regarding the molecular mechanisms through which probiotics exert their effects in the small intestine. Our study, utilizing a synthetic community of three small intestinal bacterial strains and the addition of two different probiotic species, and kynurenine as a representative dietary or endogenously produced compound, highlights the importance of selecting probiotic species with diverse genetic capabilities that complement the functional capacity of the resident microbiota, or alternatively, constructing a multispecies formula. This approach holds great promise for the development of effective probiotic therapies and underscores the need to consider the functional capacity of probiotic species when designing interventions.


Assuntos
Microbioma Gastrointestinal , Microbiota , Probióticos , Probióticos/farmacologia , Intestino Delgado , Redes e Vias Metabólicas
12.
Gut Microbes ; 15(1): 2154544, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36511640

RESUMO

Intestinal microbiota and microbiota-derived metabolites play a key role in regulating the host physiology. Recently, we have identified a gut-bacterial metabolite, namely 5-hydroxyindole, as a potent stimulant of intestinal motility via its modulation of L-type voltage-gated calcium channels located on the intestinal smooth muscle cells. Dysregulation of L-type voltage-gated calcium channels is associated with various gastrointestinal motility disorders, including constipation, making L-type voltage-gated calcium channels an important target for drug development. Nonetheless, the majority of currently available drugs are associated with alteration of the gut microbiota. Using 16S rRNA sequencing this study shows that, when administered orally, 5-hydroxyindole has only marginal effects on the rat cecal microbiota. Molecular dynamics simulations propose potential-binding pockets of 5-hydroxyindole in the α1 subunit of the L-type voltage-gated calcium channels and when its stimulatory effect on the rat colonic contractility was compared to 16 different analogues, ex-vivo, 5-hydroxyindole stood as the most potent enhancer of the intestinal contractility. Overall, the present findings imply a potential role of microbiota-derived metabolites as candidate therapeutics for targeted treatment of slow intestinal motility-related disorders including constipation.


Assuntos
Microbioma Gastrointestinal , Microbiota , Ratos , Animais , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/farmacologia , RNA Ribossômico 16S , Motilidade Gastrointestinal , Bactérias/genética , Bactérias/metabolismo , Constipação Intestinal/microbiologia
13.
Brain Behav ; 13(6): e3011, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37095714

RESUMO

INTRODUCTION: The barrier function of the gut is important for many organs and systems, including the brain. If gut permeability increases, bacterial fragments may enter the circulation, giving rise to increased systemic inflammation. Increases in bacterial translocation are reflected in higher values of blood markers, including lipopolysaccharide binding protein (LBP) and soluble cluster of differentiation 14 (sCD14). Some pioneer studies showed a negative association between bacterial translocation markers and brain volumes, but this association remains scarcely investigated. We investigate the effect of bacterial translocation on brain volumes and cognition in both healthy controls and patients with a schizophrenia spectrum disorder (SSD). MATERIALS AND METHODS: Healthy controls (n = 39) and SSD patients (n = 72) underwent an MRI-scan, venipuncture and cognition assessments. We investigated associations between LBP and sCD14 and brain volumes (intracranial volume, total brain volume, and hippocampal volume) using linear regression. We then associated LBP and sCD14 to cognitive function using a mediation analysis, with intracranial volume as mediator. RESULTS: Healthy controls showed a negative association between hippocampal volume and LBP (b = -0.11, p = .04), and intracranial volume and sCD14 (b = -0.25, p = .07). Both markers were indirectly associated with lower cognitive functioning in healthy controls (LBP: b = -0.071, p = .028; sCD14: b = -0.213, p = .052), mediated by low intracranial volume. In the SSD patients, these associations were markedly less present. CONCLUSION: These findings extend earlier studies suggesting that increased bacterial translocation may negatively affect brain volume, which indirectly impacts cognition, even in this young healthy group. If replicated, this finding stresses the importance of a healthy gut for the development and optimal functioning of the brain. Absence of these associations in the SSD group may indicate that other factors such as allostatic load, chronic medication use and interrupted educational carrier had larger impact and attenuated the relative contribution of bacterial translocation.


Assuntos
Esquizofrenia , Humanos , Esquizofrenia/diagnóstico por imagem , Receptores de Lipopolissacarídeos , Voluntários Saudáveis , Cognição , Encéfalo/diagnóstico por imagem
14.
Gut Microbes ; 15(1): 2232506, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37417553

RESUMO

The gut microbiota plays a pivotal role in health and disease. The use of probiotics as microbiota-targeted therapies is a promising strategy to improve host health. However, the molecular mechanisms involved in such therapies are often not well understood, particularly when targeting the small intestinal microbiota. In this study, we investigated the effects of a probiotic formula (Ecologic®825) on the adult human small intestinal ileostoma microbiota. The results showed that supplementation with the probiotic formula led to a reduction in the growth of pathobionts, such as Enterococcaceae and Enterobacteriaceae, and a decrease in ethanol production. These changes were associated with significant alterations in nutrient utilization and resistance to perturbations. These probiotic mediated alterations which coincided with an initial increase in lactate production and decrease in pH were followed by a sharp increase in the levels of butyrate and propionate. Moreover, the probiotic formula increased the production of multiple N-acyl amino acids in the stoma samples. The study demonstrates the utility of network theory in identifying novel microbiota-targeted therapies and improving existing ones. Overall, the findings provide insights into the dynamic molecular mechanisms underlying probiotic therapies, which can aid in the development of more effective treatments for a range of conditions.


Assuntos
Microbioma Gastrointestinal , Microbiota , Probióticos , Adulto , Humanos , Probióticos/farmacologia , Propionatos/farmacologia , Enterobacteriaceae
15.
Gut Microbes ; 14(1): 1997296, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34978524

RESUMO

The human gastrointestinal tract is home to trillions of microbes. Gut microbial communities have a significant regulatory role in the intestinal physiology, such as gut motility. Microbial effect on gut motility is often evoked by bioactive molecules from various sources, including microbial break down of carbohydrates, fibers or proteins. In turn, gut motility regulates the colonization within the microbial ecosystem. However, the underlying mechanisms of such regulation remain obscure. Deciphering the inter-regulatory mechanisms of the microbiota and bowel function is crucial for the prevention and treatment of gut dysmotility, a comorbidity associated with many diseases. In this review, we present an overview of the current knowledge on the impact of gut microbiota and its products on bowel motility. We discuss the currently available techniques employed to assess the changes in the intestinal motility. Further, we highlight the open challenges, and incorporate biophysical elements of microbes-motility interplay, in an attempt to lay the foundation for describing long-term impacts of microbial metabolite-induced changes in gut motility.


Assuntos
Bactérias/metabolismo , Microbioma Gastrointestinal , Motilidade Gastrointestinal , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/fisiologia , Animais , Bactérias/classificação , Bactérias/genética , Simulação por Computador , Humanos
16.
mSystems ; 7(1): e0119121, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35076270

RESUMO

Parkinson's disease (PD) is known to be associated with altered gastrointestinal function and microbiota composition. To date, the effect of PD medication on the gastrointestinal function and microbiota, at the site of drug absorption, the small intestine, has not been studied, although it may represent an important confounder in reported microbiota alterations observed in PD patients. To this end, healthy (non-PD) wild-type Groningen rats were employed and treated with dopamine, pramipexole (in combination with levodopa-carbidopa), or ropinirole (in combination with levodopa-carbidopa) for 14 sequential days. Rats treated with dopamine agonists showed a significant reduction in small intestinal motility and an increase in bacterial overgrowth in the distal small intestine. Notably, significant alterations in microbial taxa were observed between the treated and vehicle groups; analogous to the changes previously reported in human PD versus healthy control microbiota studies. These microbial changes included an increase in Lactobacillus and Bifidobacterium and a decrease in Lachnospiraceae and Prevotellaceae. Markedly, certain Lactobacillus species correlated negatively with levodopa levels in the systemic circulation, potentially affecting the bioavailability of levodopa. Overall, the study highlights a significant effect of PD medication intrinsically on disease-associated comorbidities, including gastrointestinal dysfunction and small intestinal bacterial overgrowth, as well as the gut microbiota composition. The results urge future studies to take into account the influence of PD medication per se when seeking to identify microbiota-related biomarkers for PD. IMPORTANCE Parkinson's disease (PD) is the second most common neurodegenerative disorder and is known to be associated with altered gastrointestinal function and microbiota composition. We previously showed that the gut bacteria harboring tyrosine decarboxylase enzymes interfere with levodopa, the main treatment for PD (S. P. van Kessel, A. K. Frye, A. O. El-Gendy, M. Castejon, A. Keshavarzian, G. van Dijk, and S. El Aidy, Nat Commun 10:310, 2019). Although PD medication could be an important confounder in the reported alterations, its effect, apart from the disease itself, on the microbiota composition or the gastrointestinal function at the site of drug absorption, the small intestine, has not been studied. The findings presented here show a significant impact of commonly prescribed PD medication on the small intestinal motility, small intestinal bacterial overgrowth, and microbiota composition, irrespective of the PD. Remarkably, we observed negative associations between bacterial species harboring tyrosine decarboxylase activity and levodopa levels in the systemic circulation, potentially affecting the bioavailability of levodopa. Overall, this study shows that PD medication is an important factor in determining gastrointestinal motility and, in turn, microbiota composition and may, partly, explain the differential abundant taxa previously reported in the cross-sectional PD microbiota human studies. The results urge future studies to take into account the influence of PD medication on gut motility and microbiota composition when seeking to identify microbiota-related biomarkers for PD.


Assuntos
Microbioma Gastrointestinal , Doença de Parkinson , Humanos , Ratos , Animais , Doença de Parkinson/tratamento farmacológico , Levodopa/uso terapêutico , Carbidopa/uso terapêutico , Tirosina Descarboxilase , Estudos Transversais , Bactérias , Motilidade Gastrointestinal
17.
Gut Microbes ; 14(1): 2081476, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35634716

RESUMO

The gut microbiota is in continuous interaction with the intestinal mucosa via metabolic, neuro-immunological, and neuroendocrine pathways. Disruption in levels of antimicrobial peptides produced by the enteroendocrine cells, such as catestatin, has been associated with changes in the gut microbiota and imbalance in intestinal homeostasis. However, whether the changes in the gut microbiota have a causational role in intestinal dyshomeostasis has remained elusive. To this end, we performed reciprocal fecal microbial transplantation in wild-type mice and mice with a knockout in the catestatin coding region of the chromogranin-A gene (CST-KO mice). Combined microbiota phylogenetic profiling, RNA sequencing, and transmission electron microscopy were employed. Fecal microbiota transplantation from mice deficient in catestatin (CST-KO) to microbiota-depleted wild-type mice induced transcriptional and physiological features characteristic of a distorted colon in the recipient animals, including impairment in tight junctions, as well as an increased collagen area fraction indicating colonic fibrosis. In contrast, fecal microbiota transplantation from wild-type mice to microbiota-depleted CST-KO mice reduced collagen fibrotic area, restored disrupted tight junction morphology, and altered fatty acid metabolism in recipient CST-KO mice. This study provides a comprehensive overview of the murine metabolic- and immune-related cellular pathways and processes that are co-mediated by the fecal microbiota transplantation and supports a prominent role for the gut microbiota in the colonic distortion associated with the lack of catestatin in mice. Overall, the data show that the gut microbiota may play a causal role in the development of features of intestinal inflammation and metabolic disorders, known to be associated with altered levels of catestatin and may, thus, provide a tractable target in the treatment and prevention of these disorders.


Assuntos
Microbioma Gastrointestinal , Transferência Adotiva , Animais , Cromogranina A , Colo , Microbioma Gastrointestinal/fisiologia , Genótipo , Camundongos , Fragmentos de Peptídeos , Fenótipo , Filogenia
18.
ISME J ; 16(8): 1873-1882, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35440728

RESUMO

The gut microbiota is in continuous interaction with the innermost layer of the gut, namely the epithelium. One of the various functions of the gut epithelium, is to keep the microbes at bay to avoid overstimulation of the underlying mucosa immune cells. To do so, the gut epithelia secrete a variety of antimicrobial peptides, such as chromogranin A (CgA) peptide catestatin (CST: hCgA352-372). As a defense mechanism, gut microbes have evolved antimicrobial resistance mechanisms to counteract the killing effect of the secreted peptides. To this end, we treated wild-type mice and CST knockout (CST-KO) mice (where only the 63 nucleotides encoding CST have been deleted) with CST for 15 consecutive days. CST treatment was associated with a shift in the diversity and composition of the microbiota in the CST-KO mice. This effect was less prominent in WT mice. Levels of the microbiota-produced short-chain fatty acids, in particular, butyrate and acetate were significantly increased in CST-treated CST-KO mice but not the WT group. Both CST-treated CST-KO and WT mice showed a significant increase in microbiota-harboring phosphoethanolamine transferase-encoding genes, which facilitate their antimicrobial resistance. Finally, we show that CST was degraded by Escherichia coli via an omptin-protease and that the abundance of this gene was significantly higher in metagenomic datasets collected from patients with Crohn's disease but not with ulcerative colitis. Overall, this study illustrates how the endogenous antimicrobial peptide, CST, shapes the microbiota composition in the gut and primes further research to uncover the role of bacterial resistance to CST in disease states such as inflammatory bowel disease.


Assuntos
Anti-Infecciosos , Microbioma Gastrointestinal , Animais , Cromogranina A/genética , Cromogranina A/metabolismo , Cromogranina A/farmacologia , Camundongos , Camundongos Knockout , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Peptídeos
19.
J Psychiatr Res ; 151: 507-515, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35636025

RESUMO

The brain-gut axis is increasingly recognized as an important contributing factor in the onset and progression of severe mental illnesses such as schizophrenia spectrum disorders and bipolar disorder. This study investigates associations between levels of faecal metabolites identified using 1H-NMR, clinical parameters, and dietary components of forty-two individuals diagnosed in a transdiagnostic approach to have severe mental illness. Faecal levels of the amino acids; alanine, leucine, and valine showed a significant positive correlation with psychiatric symptom severity as well as with dairy intake. Overall, this study proposes a diet-induced link between the brain-gut axis and the severity of psychiatric symptoms, which could be valuable in the design of novel dietary or therapeutic interventions to improve psychiatric symptoms.


Assuntos
Transtorno Bipolar , Transtornos Mentais , Esquizofrenia , Aminoácidos , Transtorno Bipolar/tratamento farmacológico , Dieta , Humanos , Transtornos Mentais/diagnóstico , Esquizofrenia/tratamento farmacológico
20.
Free Radic Biol Med ; 188: 287-297, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35753585

RESUMO

5-methoxy tryptophan (5-MTP) is an anti-fibrotic metabolite made by fibroblasts and epithelial cells, present in a micromolar concentrations in human blood, and is associated with the progression of fibrotic kidney disease, but the mechanism is unclear. Here, we show by microscopy and functional assays that 5-MTP influences mitochondria in human peripheral blood monocyte-derived macrophages. As a result, the mitochondrial membranes are more rigid, more branched, and are protected against oxidation. The macrophages also change their metabolism by reducing mitochondrial import of acyl-carnitines, intermediates of fatty acid metabolism, driving glucose import. Moreover, 5-MTP increases the endocytosis of collagen by macrophages, and experiments with inhibition of glucose uptake showed that this is a direct result of their altered metabolism. However, 5-MTP does not affect the macrophages following pathogenic stimulation, due to 5-MTP degradation by induced expression of indole-amine oxygenase-1 (IDO-1). Thus, 5-MTP is a fibrosis-protective metabolite that, in absence of pathogenic stimulation, promotes collagen uptake by anti-inflammatory macrophages by altering the physicochemical properties of their mitochondrial membranes.


Assuntos
Macrófagos , Triptofano , Colágeno/metabolismo , Fibrose , Humanos , Macrófagos/metabolismo , Mitocôndrias/metabolismo , Triptofano/metabolismo , Triptofano/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA