Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.279
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 168(5): 758-774, 2017 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-28235194

RESUMO

Because human energy metabolism evolved to favor adiposity over leanness, the availability of palatable, easily attainable, and calorically dense foods has led to unprecedented levels of obesity and its associated metabolic co-morbidities that appear resistant to traditional lifestyle interventions. However, recent progress identifying the molecular signaling pathways through which the brain and the gastrointestinal system communicate to govern energy homeostasis, combined with emerging insights on the molecular mechanisms underlying successful bariatric surgery, gives reason to be optimistic that novel precision medicines that mimic, enhance, and/or modulate gut-brain signaling can have unprecedented potential for stopping the obesity and type 2 diabetes pandemics.


Assuntos
Encéfalo/fisiologia , Metabolismo Energético , Trato Gastrointestinal/fisiologia , Animais , Regulação do Apetite , Encéfalo/anatomia & histologia , Trato Gastrointestinal/anatomia & histologia , Trato Gastrointestinal/inervação , Homeostase , Humanos , Vias Neurais , Prazer , Saciação
2.
Cell ; 168(5): 928-943.e11, 2017 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-28215708

RESUMO

Within the human gut reside diverse microbes coexisting with the host in a mutually advantageous relationship. Evidence has revealed the pivotal role of the gut microbiota in shaping the immune system. To date, only a few of these microbes have been shown to modulate specific immune parameters. Herein, we broadly identify the immunomodulatory effects of phylogenetically diverse human gut microbes. We monocolonized mice with each of 53 individual bacterial species and systematically analyzed host immunologic adaptation to colonization. Most microbes exerted several specialized, complementary, and redundant transcriptional and immunomodulatory effects. Surprisingly, these were independent of microbial phylogeny. Microbial diversity in the gut ensures robustness of the microbiota's ability to generate a consistent immunomodulatory impact, serving as a highly important epigenetic system. This study provides a foundation for investigation of gut microbiota-host mutualism, highlighting key players that could identify important therapeutics.


Assuntos
Bactérias/classificação , Microbioma Gastrointestinal , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Imunidade Adaptativa , Animais , Fenômenos Fisiológicos Bacterianos , Trato Gastrointestinal/citologia , Trato Gastrointestinal/fisiologia , Vida Livre de Germes , Humanos , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Simbiose
3.
Cell ; 161(1): 84-92, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-25815987

RESUMO

The circadian clock, a highly specialized, hierarchical network of biological pacemakers, directs and maintains proper rhythms in endocrine and metabolic pathways required for organism homeostasis. The clock adapts to environmental changes, specifically daily light-dark cycles, as well as rhythmic food intake. Nutritional challenges reprogram the clock, while time-specific food intake has been shown to have profound consequences on physiology. Importantly, a critical role in the clock-nutrition interplay appears to be played by the microbiota. The circadian clock appears to operate as a critical interface between nutrition and homeostasis, calling for more attention on the beneficial effects of chrono-nutrition.


Assuntos
Relógios Circadianos , Ingestão de Alimentos , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/fisiologia , Animais , Humanos , Metabolômica
4.
Cell ; 161(1): 12-17, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-25815980

RESUMO

Nutrition presents unanswered scientific questions of high public health importance. We envision model systems composed of interacting gastrointestinal and metabolic tissues derived from human embryonic stem cells, populated by gut microbiota. The culture will be embedded in 3D scaffolds, creating a controlled experimental system that enables tissue sampling and imaging.


Assuntos
Células-Tronco Embrionárias/citologia , Trato Gastrointestinal/fisiologia , Modelos Biológicos , Animais , Dieta , Trato Gastrointestinal/microbiologia , Humanos , Modelos Animais , Fenômenos Fisiológicos da Nutrição
5.
Cell ; 163(6): 1360-74, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26638070

RESUMO

Microbial functions in the host physiology are a result of the microbiota-host co-evolution. We show that cold exposure leads to marked shift of the microbiota composition, referred to as cold microbiota. Transplantation of the cold microbiota to germ-free mice is sufficient to increase insulin sensitivity of the host and enable tolerance to cold partly by promoting the white fat browning, leading to increased energy expenditure and fat loss. During prolonged cold, however, the body weight loss is attenuated, caused by adaptive mechanisms maximizing caloric uptake and increasing intestinal, villi, and microvilli lengths. This increased absorptive surface is transferable with the cold microbiota, leading to altered intestinal gene expression promoting tissue remodeling and suppression of apoptosis-the effect diminished by co-transplanting the most cold-downregulated strain Akkermansia muciniphila during the cold microbiota transfer. Our results demonstrate the microbiota as a key factor orchestrating the overall energy homeostasis during increased demand.


Assuntos
Metabolismo Energético , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/fisiologia , Homeostase , Tecido Adiposo Branco/metabolismo , Animais , Apoptose , Temperatura Baixa , Enterócitos/citologia , Enterócitos/metabolismo , Vida Livre de Germes , Resistência à Insulina , Absorção Intestinal , Camundongos , Verrucomicrobia/metabolismo
6.
Cell ; 163(1): 95-107, 2015 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-26406373

RESUMO

To understand how different diets, the consumers' gut microbiota, and the enteric nervous system (ENS) interact to regulate gut motility, we developed a gnotobiotic mouse model that mimics short-term dietary changes that happen when humans are traveling to places with different culinary traditions. Studying animals transplanted with the microbiota from humans representing diverse culinary traditions and fed a sequence of diets representing those of all donors, we found that correlations between bacterial species abundances and transit times are diet dependent. However, the levels of unconjugated bile acids-generated by bacterial bile salt hydrolases (BSH)-correlated with faster transit, including during consumption of a Bangladeshi diet. Mice harboring a consortium of sequenced cultured bacterial strains from the Bangladeshi donor's microbiota and fed a Bangladeshi diet revealed that the commonly used cholekinetic spice, turmeric, affects gut motility through a mechanism that reflects bacterial BSH activity and Ret signaling in the ENS. These results demonstrate how a single food ingredient interacts with a functional microbiota trait to regulate host physiology.


Assuntos
Dieta , Motilidade Gastrointestinal , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/fisiologia , Modelos Animais , Viagem , Animais , Bangladesh , Ácidos e Sais Biliares/metabolismo , Curcuma/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microbiota , Organismos Livres de Patógenos Específicos
7.
Cell ; 159(6): 1478.e1, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25480303

RESUMO

Specialized endocrine cells secrete a variety of peptide hormones all along the gastrointestinal (GI) tract, making it one of the largest endocrine organs in the body. Nutrients and developmental and neural cues trigger the secretion of gastrointestinal (GI) hormones from specialized endocrine cells along the GI tract. These hormones act in target tissues to facilitate digestion and regulate energy homeostasis. This SnapShot summarizes the production and functions of GI hormones.


Assuntos
Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/fisiologia , Hormônios/fisiologia , Animais , Trato Gastrointestinal/química , Humanos
8.
Cell ; 158(2): 300-313, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-25036630

RESUMO

Intestinal peristalsis is a dynamic physiologic process influenced by dietary and microbial changes. It is tightly regulated by complex cellular interactions; however, our understanding of these controls is incomplete. A distinct population of macrophages is distributed in the intestinal muscularis externa. We demonstrate that, in the steady state, muscularis macrophages regulate peristaltic activity of the colon. They change the pattern of smooth muscle contractions by secreting bone morphogenetic protein 2 (BMP2), which activates BMP receptor (BMPR) expressed by enteric neurons. Enteric neurons, in turn, secrete colony stimulatory factor 1 (CSF1), a growth factor required for macrophage development. Finally, stimuli from microbial commensals regulate BMP2 expression by macrophages and CSF1 expression by enteric neurons. Our findings identify a plastic, microbiota-driven crosstalk between muscularis macrophages and enteric neurons that controls gastrointestinal motility. PAPERFLICK:


Assuntos
Motilidade Gastrointestinal , Trato Gastrointestinal/citologia , Trato Gastrointestinal/microbiologia , Macrófagos/metabolismo , Animais , Proteína Morfogenética Óssea 2/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Trato Gastrointestinal/inervação , Trato Gastrointestinal/fisiologia , Técnicas In Vitro , Fator Estimulador de Colônias de Macrófagos , Camundongos , Neurônios/metabolismo , Peristaltismo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Transdução de Sinais
9.
Cell ; 153(4): 797-811, 2013 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-23663779

RESUMO

All metazoan guts are subjected to immunologically unique conditions in which an efficient antimicrobial system operates to eliminate pathogens while tolerating symbiotic commensal microbiota. However, the molecular mechanisms controlling this process are only partially understood. Here, we show that bacterial-derived uracil acts as a ligand for dual oxidase (DUOX)-dependent reactive oxygen species generation in Drosophila gut and that the uracil production in bacteria causes inflammation in the gut. The acute and controlled uracil-induced immune response is required for efficient elimination of bacteria, intestinal cell repair, and host survival during infection of nonresident species. Among resident gut microbiota, uracil production is absent in symbionts, allowing harmonious colonization without DUOX activation, whereas uracil release from opportunistic pathobionts provokes chronic inflammation. These results reveal that bacteria with distinct abilities to activate uracil-induced gut inflammation, in terms of intensity and duration, act as critical factors that determine homeostasis or pathogenesis in gut-microbe interactions.


Assuntos
Drosophila/imunologia , Drosophila/microbiologia , Imunidade nas Mucosas , Pectobacterium carotovorum/fisiologia , Simbiose , Uracila/metabolismo , Animais , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/fisiologia , Homeostase , Humanos , Inflamação/imunologia , Inflamação/microbiologia , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/microbiologia , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco/metabolismo
10.
Nature ; 606(7912): 94-101, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35650358

RESUMO

Neurotransmitters play essential roles in regulating neural circuit dynamics both in the central nervous system as well as at the peripheral, including the gastrointestinal tract1-3. Their real-time monitoring will offer critical information for understanding neural function and diagnosing disease1-3. However, bioelectronic tools to monitor the dynamics of neurotransmitters in vivo, especially in the enteric nervous systems, are underdeveloped. This is mainly owing to the limited availability of biosensing tools that are capable of examining soft, complex and actively moving organs. Here we introduce a tissue-mimicking, stretchable, neurochemical biological interface termed NeuroString, which is prepared by laser patterning of a metal-complexed polyimide into an interconnected graphene/nanoparticle network embedded in an elastomer. NeuroString sensors allow chronic in vivo real-time, multichannel and multiplexed monoamine sensing in the brain of behaving mouse, as well as measuring serotonin dynamics in the gut without undesired stimulations and perturbing peristaltic movements. The described elastic and conformable biosensing interface has broad potential for studying the impact of neurotransmitters on gut microbes, brain-gut communication and may ultimately be extended to biomolecular sensing in other soft organs across the body.


Assuntos
Encéfalo , Sistema Nervoso Entérico , Trato Gastrointestinal , Neurotransmissores , Animais , Técnicas Biossensoriais , Encéfalo/metabolismo , Eixo Encéfalo-Intestino , Elastômeros , Sistema Nervoso Entérico/metabolismo , Trato Gastrointestinal/inervação , Trato Gastrointestinal/fisiologia , Grafite , Lasers , Camundongos , Nanopartículas , Neurotransmissores/análise , Serotonina/análise
11.
Cell ; 151(5): 1113-25, 2012 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-23178127

RESUMO

Internal nutrient sensors play important roles in feeding behavior, yet their molecular structure and mechanism of action are poorly understood. Using Ca(2+) imaging and behavioral assays, we show that the gustatory receptor 43a (Gr43a) functions as a narrowly tuned fructose receptor in taste neurons. Remarkably, Gr43a also functions as a fructose receptor in the brain. Interestingly, hemolymph fructose levels are tightly linked to feeding status: after nutritious carbohydrate consumption, fructose levels rise several fold and reach a concentration sufficient to activate Gr43a in the brain. By using different feeding paradigms and artificial activation of Gr43a-expressing brain neurons, we show that Gr43a is both necessary and sufficient to sense hemolymph fructose and promote feeding in hungry flies but suppress feeding in satiated flies. Thus, our studies indicate that the Gr43a-expressing brain neurons function as a nutrient sensor for hemolymph fructose and assign opposing valence to feeding experiences in a satiation-dependent manner.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/fisiologia , Frutose/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Encéfalo/fisiologia , Alimentos , Trato Gastrointestinal/fisiologia , Hemolinfa/metabolismo , Fome , Neurônios/metabolismo , Resposta de Saciedade
12.
Proc Natl Acad Sci U S A ; 121(28): e2310992121, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38968105

RESUMO

Tissue buckling is an increasingly appreciated mode of morphogenesis in the embryo, but it is often unclear how geometric and material parameters are molecularly determined in native developmental contexts to generate diverse functional patterns. Here, we study the link between differential mechanical properties and the morphogenesis of distinct anteroposterior compartments in the intestinal tract-the esophagus, small intestine, and large intestine. These regions originate from a simple, common tube but adopt unique forms. Using measured data from the developing chick gut coupled with a minimal theory and simulations of differential growth, we investigate divergent lumen morphologies along the entire early gut and demonstrate that spatiotemporal geometries, moduli, and growth rates control the segment-specific patterns of mucosal buckling. Primary buckling into wrinkles, folds, and creases along the gut, as well as secondary buckling phenomena, including period-doubling in the foregut and multiscale creasing-wrinkling in the hindgut, are captured and well explained by mechanical models. This study advances our existing knowledge of how identity leads to form in these regions, laying the foundation for future work uncovering the relationship between molecules and mechanics in gut morphological regionalization.


Assuntos
Morfogênese , Animais , Embrião de Galinha , Morfogênese/fisiologia , Fenômenos Biomecânicos , Galinhas , Trato Gastrointestinal/fisiologia , Trato Gastrointestinal/anatomia & histologia , Modelos Biológicos , Intestinos/fisiologia , Intestinos/embriologia
13.
Immunity ; 46(6): 910-926, 2017 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-28636959

RESUMO

Interactions between the nervous and immune systems enable the gut to respond to the variety of dietary products that it absorbs, the broad spectrum of pathogens that it encounters, and the diverse microbiome that it harbors. The enteric nervous system (ENS) senses and reacts to the dynamic ecosystem of the gastrointestinal (GI) tract by translating chemical cues from the environment into neuronal impulses that propagate throughout the gut and into other organs in the body, including the central nervous system (CNS). This review will describe the current understanding of the anatomy and physiology of the GI tract by focusing on the ENS and the mucosal immune system. We highlight emerging literature that the ENS is essential for important aspects of microbe-induced immune responses in the gut. Although most basic and applied research in neuroscience has focused on the brain, the proximity of the ENS to the immune system and its interface with the external environment suggest that novel paradigms for nervous system function await discovery.


Assuntos
Sistema Nervoso Central/imunologia , Sistema Nervoso Entérico , Microbioma Gastrointestinal , Trato Gastrointestinal/fisiologia , Sistema Imunitário/imunologia , Imunidade nas Mucosas , Intestinos/imunologia , Animais , Exposição Ambiental , Trato Gastrointestinal/anatomia & histologia , Interações Hospedeiro-Patógeno , Humanos , Neuroimunomodulação
14.
Annu Rev Neurosci ; 40: 21-49, 2017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28301775

RESUMO

The microbiota is increasingly recognized for its ability to influence the development and function of the nervous system and several complex host behaviors. In this review, we discuss emerging roles for the gut microbiota in modulating host social and communicative behavior, stressor-induced behavior, and performance in learning and memory tasks. We summarize effects of the microbiota on host neurophysiology, including brain microstructure, gene expression, and neurochemical metabolism across regions of the amygdala, hippocampus, frontal cortex, and hypothalamus. We further assess evidence linking dysbiosis of the gut microbiota to neurobehavioral diseases, such as autism spectrum disorder and major depression, drawing upon findings from animal models and human trials. Finally, based on increasing associations between the microbiota, neurophysiology, and behavior, we consider whether investigating mechanisms underlying the microbiota-gut-brain axis could lead to novel approaches for treating particular neurological conditions.


Assuntos
Encéfalo/fisiologia , Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/fisiologia , Transtornos Mentais/fisiopatologia , Animais , Humanos , Transtornos Mentais/microbiologia
15.
Nature ; 568(7750): 98-102, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30918408

RESUMO

Satiation is the process by which eating and drinking reduce appetite. For thirst, oropharyngeal cues have a critical role in driving satiation by reporting to the brain the volume of fluid that has been ingested1-12. By contrast, the mechanisms that relay the osmolarity of ingested fluids remain poorly understood. Here we show that the water and salt content of the gastrointestinal tract are precisely measured and then rapidly communicated to the brain to control drinking behaviour in mice. We demonstrate that this osmosensory signal is necessary and sufficient for satiation during normal drinking, involves the vagus nerve and is transmitted to key forebrain neurons that control thirst and vasopressin secretion. Using microendoscopic imaging, we show that individual neurons compute homeostatic need by integrating this gastrointestinal osmosensory information with oropharyngeal and blood-borne signals. These findings reveal how the fluid homeostasis system monitors the osmolarity of ingested fluids to dynamically control drinking behaviour.


Assuntos
Encéfalo/fisiologia , Ingestão de Líquidos/fisiologia , Trato Gastrointestinal/fisiologia , Neurônios/fisiologia , Saciação/fisiologia , Sede/fisiologia , Animais , Encéfalo/citologia , Feminino , Neurônios GABAérgicos/metabolismo , Trato Gastrointestinal/inervação , Glutamatos/metabolismo , Masculino , Camundongos , Orofaringe/inervação , Orofaringe/fisiologia , Concentração Osmolar , Prosencéfalo/metabolismo , Nervo Vago/fisiologia , Vasopressinas/metabolismo
16.
Am J Physiol Gastrointest Liver Physiol ; 326(6): G712-G725, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38626403

RESUMO

Gut physiology is the epicenter of a web of internal communication systems (i.e., neural, immune, hormonal) mediated by cell-cell contacts, soluble factors, and external influences, such as the microbiome, diet, and the physical environment. Together these provide the signals that shape enteric homeostasis and, when they go awry, lead to disease. Faced with the seemingly paradoxical tasks of nutrient uptake (digestion) and retarding pathogen invasion (host defense), the gut integrates interactions between a variety of cells and signaling molecules to keep the host nourished and protected from pathogens. When the system fails, the outcome can be acute or chronic disease, often labeled as "idiopathic" in nature (e.g., irritable bowel syndrome, inflammatory bowel disease). Here we underscore the importance of a holistic approach to gut physiology, placing an emphasis on intercellular connectedness, using enteric neuroimmunophysiology as the paradigm. The goal of this opinion piece is to acknowledge the pace of change brought to our field via single-cell and -omic methodologies and other techniques such as cell lineage tracing, transgenic animal models, methods for culturing patient tissue, and advanced imaging. We identify gaps in the field and hope to inspire and challenge colleagues to take up the mantle and advance awareness of the subtleties, intricacies, and nuances of intestinal physiology in health and disease by defining communication pathways between gut resident cells, those recruited from the circulation, and "external" influences such as the central nervous system and the gut microbiota.


Assuntos
Microbioma Gastrointestinal , Trato Gastrointestinal , Humanos , Animais , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/fisiologia , Trato Gastrointestinal/microbiologia , Microbioma Gastrointestinal/fisiologia , Neuroimunomodulação/fisiologia , Sistema Nervoso Entérico/fisiologia , Sistema Nervoso Entérico/imunologia
17.
Mol Pharm ; 21(8): 3824-3837, 2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-38958668

RESUMO

In vivo studies of formulation performance with in vitro and/or in silico simulations are often limited by significant gaps in our knowledge of the interaction between administered dosage forms and the human gastrointestinal tract. This work presents a novel approach for the investigation of gastric motility influence on dosage form performance, by combining biopredictive dissolution tests in an innovative PhysioCell apparatus with mechanistic physiology-based pharmacokinetic modeling. The methodology was based on the pharmacokinetic data from a large (n = 118) cohort of healthy volunteers who ingested a capsule containing a highly soluble and rapidly absorbed drug under fasted conditions. The developed dissolution tests included biorelevant media, varied fluid flows, and mechanical stress events of physiological timing and intensity. The dissolution results were used as inputs for pharmacokinetic modeling that led to the deduction of five patterns of gastric motility and their prevalence in the studied population. As these patterns significantly influenced the observed pharmacokinetic profiles, the proposed methodology is potentially useful to other in vitro-in vivo predictions involving immediate-release oral dosage forms.


Assuntos
Motilidade Gastrointestinal , Solubilidade , Humanos , Motilidade Gastrointestinal/fisiologia , Adulto , Masculino , Feminino , Modelos Biológicos , Administração Oral , Adulto Jovem , Voluntários Saudáveis , Simulação por Computador , Liberação Controlada de Fármacos/fisiologia , Pessoa de Meia-Idade , Jejum/fisiologia , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/fisiologia
18.
Curr Opin Crit Care ; 30(4): 290-297, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38872371

RESUMO

PURPOSE OF REVIEW: Gastrointestinal (GI) dysfunction limits enteral nutrition (EN) delivery in critical illness and contributes to systemic inflammation. The enteroendocrine (EE) axis plays an integral role in this interface between nutrition, inflammation, and GI function in critical illness. In this review, we present an overview of the EE system with a focus on its role in GI inflammation and function. RECENT FINDINGS: Enteroendocrine cells have been primarily described in their role in macronutrient digestion and absorption. Recent research has expanded on the diverse functions of EE cells including their ability to sense microbial peptides and metabolites and regulate immune function and inflammation. Therefore, EE cells may be both affected by and contribute to many pathophysiologic states and interventions of critical illness such as dysbiosis , inflammation, and alternative EN strategies. In this review, we present an overview of EE cells including their growing role in nonnutrient functions and integrate this understanding into relevant aspects of critical illness with a focus on EN. SUMMARY: The EE system is key in maintaining GI homeostasis in critical illness, and how it is impacted and contributes to outcomes in the setting of dysbiosis , inflammation and different feeding strategies in critical illness should be considered.


Assuntos
Estado Terminal , Nutrição Enteral , Células Enteroendócrinas , Inflamação , Humanos , Inflamação/fisiopatologia , Células Enteroendócrinas/fisiologia , Disbiose/fisiopatologia , Trato Gastrointestinal/fisiopatologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/fisiologia , Microbioma Gastrointestinal/fisiologia , Gastroenteropatias/fisiopatologia , Estado Nutricional/fisiologia
19.
Eur J Appl Physiol ; 124(5): 1601-1608, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38172425

RESUMO

PURPOSE: We determined the effects of different environmental temperatures on exercise-induced gastrointestinal (GI) damage and delayed gastric emptying (GE) rate. METHODS: Eleven trained males completed three trials on different days, consisting of (1) exercise in a thermoneutral environment (CON, 23 °C), (2) exercise in a hot environment (HOT, 35 °C), and (3) exercise in a cold environment (COLD, 10 °C). The subjects performed high-intensity interval-type endurance exercises in all trials. Blood intestinal fatty acid binding protein (I-FABP) levels was determine before and after exercise. We evaluated Tmax (time when the 13C-excretion/h reached a maximum level) as an indication of the GE rate during post-exercise. RESULTS: Rectal temperature during exercise was significantly higher (P < 0.001) in the HOT (38.7 ± 0.3 °C) trial compared with the CON (38.2 ± 0.3 °C) and COLD (38.2 ± 0.3 °C) trials, with no significant difference between the CON and COLD trials. Plasma I-FABP level after exercise (relative to the pre-exercise level) were significantly greater (P = 0.005) in the HOT trial (92.9 ± 69.6%) than in the CON (37.2 ± 31.6%) and COLD (37.6 ± 41.8%) trials. However, there was no significant difference between the CON and COLD trials. Moreover, the Tmax was delayed significantly (P = 0.006) in the HOT trial compared with the CON and COLD trials, with no significant difference between the CON and COLD trials. CONCLUSION: GI function following endurance exercise was similar between thermoneutral and cold environments, while endurance exercise in a hot environment exacerbated GI function compared with thermoneutral and cold environments.


Assuntos
Exercício Físico , Proteínas de Ligação a Ácido Graxo , Resistência Física , Humanos , Masculino , Proteínas de Ligação a Ácido Graxo/sangue , Resistência Física/fisiologia , Exercício Físico/fisiologia , Adulto , Temperatura Baixa , Esvaziamento Gástrico/fisiologia , Trato Gastrointestinal/fisiologia , Temperatura Alta , Adulto Jovem , Temperatura Corporal/fisiologia
20.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33853946

RESUMO

Animal gastrointestinal tracts harbor a microbiome that is integral to host function, yet species from diverse phyla have evolved a reduced digestive system or lost it completely. Whether such changes are associated with alterations in the diversity and/or abundance of the microbiome remains an untested hypothesis in evolutionary symbiosis. Here, using the life history transition from planktotrophy (feeding) to lecithotrophy (nonfeeding) in the sea urchin Heliocidaris, we demonstrate that the lack of a functional gut corresponds with a reduction in microbial community diversity and abundance as well as the association with a diet-specific microbiome. We also determine that the lecithotroph vertically transmits a Rickettsiales that may complement host nutrition through amino acid biosynthesis and influence host reproduction. Our results indicate that the evolutionary loss of a functional gut correlates with a reduction in the microbiome and the association with an endosymbiont. Symbiotic transitions can therefore accompany life history transitions in the evolution of developmental strategies.


Assuntos
Trato Gastrointestinal/microbiologia , Ouriços-do-Mar/microbiologia , Simbiose/genética , Adaptação Biológica/genética , Animais , Evolução Biológica , Trato Gastrointestinal/fisiologia , Microbiota/genética , Filogenia , RNA Ribossômico 16S/genética , Ouriços-do-Mar/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA