Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 397
1.
Article En | MEDLINE | ID: mdl-34737157

Prominent ontogenetic changes of the gastrointestinal tract (GIT) should occur in mammals whose neonatal diet of milk differs from that of adults, and especially in herbivores (as vegetation is particularly distinct from milk), and even more so in foregut fermenters, whose forestomach only becomes functionally relevant with vegetation intake. Due to the protracted lactation in marsupials, ontogenetic differences can be particularly well investigated in this group. Here, we report body mass (BM) scaling relationships of wet GIT content mass in 28 in-pouch young (50 g to 3 kg) and 15 adult (16-70 kg) western grey kangaroos Macropus fuliginosus melanops. Apart from the small intestinal contents, in-pouch young and adults did not differ in the scaling exponents ('slope' in log-log plots) but did differ in the scaling factor ('intercept'), with an implied substantial increase in wet GIT content mass during the out-of-pouch juvenile period. In contrast to forestomach contents, caecum contents were elevated in juveniles still in the pouch, suggestive of fermentative digestion of milk and intestinal secretion residues, particularly in the caecum. The substantial increase in GIT contents (from less than 1 to 10-20% of BM) was associated mainly with the increase in forestomach contents (from 25 to 80% of total GIT contents) and a concomitant decrease in small intestine contents (from 50 to 8%), emphasizing the shifting relevance of auto-enzymatic and allo-enzymatic (microbial) digestion. There was a concomitant increase in the contents-to-tissue ratio of the fermentation chambers (forestomach and caecum), but this ratio generally did not change for the small intestine. Our study not only documents significant ontogenetic changes in digestive morpho-physiology, but also exemplifies the usefulness of intraspecific allometric analyses for quantifying these changes.


Gastrointestinal Contents/chemistry , Gastrointestinal Tract/growth & development , Gastrointestinal Tract/physiology , Macropodidae/growth & development , Macropodidae/physiology , Animals , Digestive System Physiological Phenomena , Female , Fermentation/physiology , Male , Models, Biological
2.
Cell Mol Life Sci ; 79(1): 46, 2021 Dec 22.
Article En | MEDLINE | ID: mdl-34936034

Gastrointestinal (GI) problems and microbiota alterations have been frequently reported in autism spectrum disorders (ASD). In addition, abnormal perinatal trace metal levels have been found in ASD. Accordingly, mice exposed to prenatal zinc deficiency display features of ASD-like behavior. Here, we model GI development using 3D intestinal organoids grown under zinc-restricted conditions. We found significant morphological alterations. Using proteomic approaches, we identified biological processes affected by zinc deficiency that regulate barrier permeability and pro-inflammatory pathways. We confirmed our results in vivo through proteomics studies and investigating GI development in zinc-deficient mice. These show altered GI physiology and pro-inflammatory signaling, resulting in chronic systemic and neuroinflammation, and gut microbiota composition similar to that reported in human ASD cases. Thus, low zinc status during development is sufficient to compromise intestinal barrier integrity and activate pro-inflammatory signaling, resulting in changes in microbiota composition that may aggravate inflammation, altogether mimicking the co-morbidities frequently observed in ASD.


Autism Spectrum Disorder , Gastrointestinal Diseases , Neuroinflammatory Diseases , Zinc/deficiency , Animals , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/microbiology , Female , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/microbiology , Gastrointestinal Microbiome , Gastrointestinal Tract/growth & development , Male , Mice , Mice, Inbred C57BL , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/microbiology , Organoids , Proteomics
3.
Development ; 148(21)2021 11 01.
Article En | MEDLINE | ID: mdl-34758081

The developmental programs that build and sustain animal forms also encode the capacity to sense and adapt to the microbial world within which they evolved. This is abundantly apparent in the development of the digestive tract, which typically harbors the densest microbial communities of the body. Here, we review studies in human, mouse, zebrafish and Drosophila that are revealing how the microbiota impacts the development of the gut and its communication with the nervous system, highlighting important implications for human and animal health.


Brain-Gut Axis/physiology , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/growth & development , Animals , Cell Lineage , Enteric Nervous System/cytology , Enteric Nervous System/growth & development , Enteric Nervous System/physiology , Gastrointestinal Motility , Gastrointestinal Tract/innervation , Gastrointestinal Tract/microbiology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/growth & development , Neurons/cytology , Neurons/physiology
4.
Elife ; 102021 09 23.
Article En | MEDLINE | ID: mdl-34553686

The gut is the primary interface between an animal and food, but how it adapts to qualitative dietary variation is poorly defined. We find that the Drosophila midgut plastically resizes following changes in dietary composition. A panel of nutrients collectively promote gut growth, which sugar opposes. Diet influences absolute and relative levels of enterocyte loss and stem cell proliferation, which together determine cell numbers. Diet also influences enterocyte size. A high sugar diet inhibits translation and uncouples intestinal stem cell proliferation from expression of niche-derived signals, but, surprisingly, rescuing these effects genetically was not sufficient to modify diet's impact on midgut size. However, when stem cell proliferation was deficient, diet's impact on enterocyte size was enhanced, and reducing enterocyte-autonomous TOR signaling was sufficient to attenuate diet-dependent midgut resizing. These data clarify the complex relationships between nutrition, epithelial dynamics, and cell size, and reveal a new mode of plastic, diet-dependent organ resizing.


Diet , Drosophila melanogaster/growth & development , Gastrointestinal Tract/growth & development , Animals , Animals, Genetically Modified , Cell Proliferation , Drosophila melanogaster/physiology , Enterocytes/cytology , Female , Gastrointestinal Tract/cytology , Gastrointestinal Tract/physiology , Male , Stem Cell Niche
5.
Genes (Basel) ; 12(8)2021 08 23.
Article En | MEDLINE | ID: mdl-34440466

Mesodermal cells of holothurian Eupentacta fraudatrix can transdifferentiate into enterocytes during the regeneration of the digestive system. In this study, we investigated the expression of several genes involved in gut regeneration in E. fraudatrix. Moreover, the localization of progenitor cells of coelomocytes, juvenile cells, and their participation in the formation of the luminal epithelium of the digestive tube were studied. It was shown that Piwi-positive cells were not involved in the formation of the luminal epithelium of the digestive tube. Ef-72 kDa type IV collagenase and Ef-MMP16 had an individual expression profile and possibly different functions. The Ef-tensilin3 gene exhibited the highest expression and indicates its potential role in regeneration. Ef-Sox9/10 and Ef-Sox17 in E. fraudatrix may participate in the mechanism of transdifferentiation of coelomic epithelial cells. Their transcripts mark the cells that plunge into the connective tissue of the gut anlage and give rise to enterocytes. Ef-Sox9/10 probably controls the switching of mesodermal cells to the enterocyte phenotype, while Ef-Sox17 may be involved in the regulation of the initial stages of transdifferentiation.


Digestive System/growth & development , Gastrointestinal Tract/growth & development , Regeneration/genetics , Sea Cucumbers/genetics , Animals , Cell Transdifferentiation/genetics , Digestive System/metabolism , Epithelial Cells/metabolism , Gastrointestinal Tract/metabolism , Gene Expression Regulation, Developmental/genetics , Matrix Metalloproteinases/genetics , Mesoderm/growth & development , Mesoderm/metabolism , RNA, Small Interfering/genetics , SOX Transcription Factors/genetics , Sea Cucumbers/growth & development , Tissue Inhibitor of Metalloproteinases/genetics
6.
J Insect Physiol ; 133: 104274, 2021.
Article En | MEDLINE | ID: mdl-34216600

BACKGROUND: Digestive tissues are essential for diet processing and nutrient accessibility, especially in omnivores, and these functions occur despite and in collaboration with dynamic microbial communities that reside within and upon these tissues. Prolonged host development and reduced digestive tissue sizes have been observed in germ-free animals, and normal host phenotypes were recovered following the re-introduction of typical gut microbiomes via coprophagy. RESULTS: High-resolution histological analyses of Periplaneta americana cockroach digestive tissues revealed that total prevention of microbial colonization of the gut had severe impacts on the growth and development of gut tissues, especially the posterior midgut and anterior hindgut subcompartments that are expected to be colonized and inhabited by the greatest number of bacteria. Juveniles that were briefly exposed to normal gut microbiota exhibited a partial gut morphological recovery, suggesting that a single inoculation was insufficient. These data highlight gut microbiota as integral to normal growth and development of tissues they are in direct contact with and, more broadly, the organism in which they reside. CONCLUSIONS: We draw on these data, host life history traits (i.e. multigenerational cohousing, molting, and filial coprophagy and exuvia feeding), and previous studies to suggest a host developmental model in which gut tissues reflect a conflict-collaboration dynamic where 1) nutrient-absorptive anterior midgut tissues are in competition with transient and resident bacteria for easily assimilable dietary nutrients and whose growth is least-affected by the presence of gut bacteria and 2) posterior midgut, anterior hindgut, and to a lesser degree, posterior hindgut tissues are significantly impacted by gut bacterial presence because they are occupied by the greatest number of bacteria and the host is relying upon, and thus collaborating with, them to assist with complex polysaccharide catabolism processing and nutrient provisioning (i.e. short-chain fatty acids).


Gastrointestinal Microbiome , Periplaneta/growth & development , Periplaneta/microbiology , Animals , Gastrointestinal Tract/growth & development , Gastrointestinal Tract/microbiology , Nymph/growth & development , Nymph/microbiology
7.
Food Funct ; 12(17): 8007-8016, 2021 Sep 07.
Article En | MEDLINE | ID: mdl-34269359

Kiwifruit (KF) fiber, a mixture of soluble and insoluble fibers, elicits mucosal changes in the gastrointestinal tract (GIT). This study aimed to define the nature of these changes in mucosal features throughout the GIT of the growing pig in response to semi-synthetic iso-fiber diets containing cellulose (CEL, low GIT luminal functionality) as the sole fiber source (4.5%), or diets where half of the CEL was replaced by either PSY fiber (PSY husk, high GIT luminal functionality) or KF fiber (consumed as intact fruit). Entire male growing pigs (n = 24, 21 kg bodyweight) received the three diets (n = 8) for 42 d. GIT tissues, digesta, and feces were sampled. The partial replacement of CEL increased (P≤ 0.05) the ileal (KF 22% and PSY 33%) and colonic (PSY 86%) mucus layer thickness, whereas it decreased the rectal crypt depth (KF -26%), and small intestinal (duodenum to ileum) villus length (PSY -17%). The number of duodenal goblet cells was 77% higher (P≤ 0.05) for KF than CEL. Pigs fed the KF-containing diet had greater (P≤ 0.05) apparent ileal organic matter digestibility and apparent total tract organic matter digestibility compared with CEL, but the lowest amount of fermented organic matter in the large intestine. In conclusion, partial substitution of CEL with PSY or KF at a constant, practically-relevant dietary fiber intake, affected several measures of GIT functionality with effects being specific to the added fiber.


Actinidia/metabolism , Cellulose/metabolism , Gastrointestinal Tract/metabolism , Mucus/metabolism , Psyllium/metabolism , Swine/metabolism , Animal Feed/analysis , Animals , Dietary Fiber/metabolism , Digestion , Fruit/metabolism , Gastrointestinal Tract/growth & development , Histology , Male , Swine/growth & development
8.
Sci Rep ; 11(1): 12808, 2021 06 17.
Article En | MEDLINE | ID: mdl-34140588

Functionality of the gastrointestinal tract is essential for growth and development of newborns. Preterm infants have an immature gastrointestinal tract, which is a major challenge in neonatal care. This study aims to improve the understanding of gastrointestinal functionality and maturation during the early life of preterm infants by means of gastrointestinal enzyme activity assays and metaproteomics. In this single-center, observational study, preterm infants born between 24 and 33 weeks (n = 40) and term infants born between 37 and 42 weeks (n = 3), who were admitted to Isala (Zwolle, the Netherlands), were studied. Enzyme activity analyses identified active proteases in gastric aspirates of preterm infants. Metaproteomics revealed human milk, digestive and immunological proteins in gastric aspirates of preterm infants and feces of preterm and term infants. The fecal proteome of preterm infants was deprived of gastrointestinal barrier-related proteins during the first six postnatal weeks compared to term infants. In preterm infants, bacterial oxidative stress proteins were increased compared to term infants and higher birth weight correlated to higher relative abundance of bifidobacterial proteins in postnatal week 3 to 6. Our findings indicate that gastrointestinal and beneficial microbial proteins involved in gastrointestinal maturity are associated with gestational and postnatal age.


Bacteria/metabolism , Biomarkers/metabolism , Digestion/physiology , Gastrointestinal Tract/growth & development , Gastrointestinal Tract/microbiology , Infant, Premature/physiology , Animals , Cattle , Gastrointestinal Tract/enzymology , Humans , Hydrogen-Ion Concentration , Infant, Newborn , Milk Proteins/metabolism , Oxidative Stress , Peptide Hydrolases/metabolism , Proteome/metabolism , Proteomics , Time Factors
9.
Dev Cell ; 56(14): 2059-2072.e3, 2021 07 26.
Article En | MEDLINE | ID: mdl-34019841

Individual organ development must be temporally coordinated with development of the rest of the organism. As a result, cell division cycles in a developing organ occur on a relatively fixed timescale. Despite this, many developing organs can regenerate cells lost to injury. How organs regenerate within the time constraints of organism development remains unclear. Here, we show that the developing Drosophila hindgut regenerates by accelerating the mitotic cell cycle. This process is achieved by decreasing G1 length and requires the JAK/STAT ligand unpaired-3. Mitotic capacity is then terminated by the steroid hormone ecdysone receptor and the Sox transcription factor Dichaete. These two factors converge on regulation of a hindgut-specific enhancer of fizzy-related, a negative regulator of mitotic cyclins. Our findings reveal how the cell-cycle machinery and cytokine signaling can be adapted to accomplish developmental organ regeneration.


Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , G1 Phase , Gastrointestinal Tract/growth & development , Gene Expression Regulation, Developmental , Organogenesis , Regeneration , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , Gastrointestinal Tract/injuries , Gastrointestinal Tract/metabolism , Janus Kinases/genetics , Janus Kinases/metabolism , Male , Mitosis , SOX Transcription Factors/genetics , SOX Transcription Factors/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Exp Cell Res ; 402(2): 112574, 2021 05 15.
Article En | MEDLINE | ID: mdl-33794264

Congenital anorectal malformations (ARMs) are among the most prominent deformities of the gastrointestinal tract; however, their precise aetiology remains obscure. Immunohistochemistry demonstrated that, in the ARM group, the PPPDE1-positive cells were widely distributed in the hindgut epithelial tissue from GD13 to GD16. Immunofluorescence revealed that most TUNEL-, Bax-, and Cytochrome C (Cyt C)-positive cells overlapped with PPPDE1-positive cells in the urorectal septum (URS). Western blotting and quantitative real-time RT-PCR revealed that PPPDE1 levels were significantly higher in the ARM group from GD13 to GD14 (p < 0.05). IEC-6 cells were transfected with PPPDE1 overexpression plasmid/NC (negative control) or si-PPPDE1/si-NC. Flow cytometry analysis and CCK-8 assay (used to detect apoptosis and proliferation, respectively), as well as western blotting, showed that the levels of PPPDE1 were positively correlated with the pro-apoptotic molecules Bax and Cyt C. Accordingly, aberrantly high expression of PPPDE1 caused a spatiotemporal imbalance in foetal rats with ARMs during hindgut development. Therefore, the upregulation of PPPDE1 may promote epithelial apoptosis and reduce proliferation in the hindgut via the mitochondrial apoptotic pathway. This could affect the fusion of the URS and cloacal membrane, ultimately inhibiting the hindgut development and resulting in ARMs.


Anorectal Malformations/genetics , Carbon-Nitrogen Lyases/genetics , Gastrointestinal Tract/metabolism , bcl-2-Associated X Protein/genetics , Animals , Anorectal Malformations/pathology , Apoptosis/genetics , Cell Proliferation/genetics , Cytochromes c/genetics , Embryo, Mammalian , Fetal Development/genetics , Gastrointestinal Tract/growth & development , Gastrointestinal Tract/pathology , Humans , Mitochondria/genetics , Rats , Signal Transduction/genetics , Transcriptional Activation/genetics
11.
Zoology (Jena) ; 146: 125905, 2021 06.
Article En | MEDLINE | ID: mdl-33631602

In vertebrates, gut coiling proceeds left-right asymmetrically during expansion of the gastrointestinal tract with highly organized muscular structures facilitating peristalsis. In this report, we explored the mechanisms of larval gut coiling morphogenesis relevant to its nascent smooth muscle cells using highly transparent Xenopus early larvae. First, to visualize the dynamics of intestinal smooth muscle cells, whole-mount specimens were immunostained with anti-smooth muscle-specific actin (SM-actin) antibody. We found that the nascent gut of Xenopus early larvae gradually expands the SM-actin-positive region in a stage-dependent manner. Transverse orientation of smooth muscle cells was first established, and next, the cellular longitudinal orientation along the gut axis was followed to make a meshwork of the contractile cells. Finally, anisotropic torsion by the smooth muscle cells was generated in the center of gut coiling, suggesting that twisting force might be involved in the late phase of coiling morphogenesis of the gut. Administration of S-(-)-Blebbistatin to attenuate the actomyosin contraction in vivo resulted in cancellation of coiling of the gut. Development of decapitation embryos, trunk 'torso' explants, and gut-only explants revealed that initial coiling of the gut proceeds without interactions with the other parts of the body including the central nervous system. We newly developed an in vitro model to assess the gut coiling morphogenesis, indicating that coiling pattern of the nascent Xenopus gut is partially gut-autonomous. Using this gut explant culture technique, inhibition of actomyosin contraction was performed by administrating either actin polymerization inhibitor, myosin light chain kinase inhibitor, or calmodulin antagonist. All of these reagents decreased the extent of gut coiling morphogenesis in vitro. Taken together, these results suggest that the contraction force generated by actomyosin-rich intestinal smooth muscle cells during larval stages is essential for the normal coiling morphogenesis of this muscular tubular organ.


Gastrointestinal Tract/growth & development , Muscle Development/physiology , Myocytes, Smooth Muscle/physiology , Xenopus laevis/growth & development , Animals , Larva
12.
Poult Sci ; 100(2): 1205-1212, 2021 Feb.
Article En | MEDLINE | ID: mdl-33518078

Sustainability of poultry farming relies on the development of more efficient and autonomous production systems in terms of feed supply. This implies a better integration of adaptive traits in breeding programs, including digestive efficiency, to favor the use of a wider variety of feedstuffs. The objective of the study was to better characterize the kinetics of development of the digestive tract in broilers, in relationship with digestive efficiency by measuring various digestive parameters as well as serum color. Absolute and relative growth of gastrointestinal tract organs were compared between 2 divergent chicken lines selected on digestive efficiency (AMEn) during 7 wk of development. We show that as early as 7 d of age, these 2 lines differs for several organs developments and that these differences remain visible later on. In addition, the allometry of the gizzard and intestine segments is different between the 2 lines, with efficient birds putting more effort in the upper part of the digestive tract during postnatal development and less-efficient birds putting more effort in the lower part of the gastrointestinal tract. Interestingly, we also showed that differences in serum pigmentation, which is a good biomarker for digestive capacity, could be a convenient diagnostic tool to discriminate between chickens with high or low digestive efficiency at early stages of development. In conclusion, this study showed that selection of chickens for AMEn had large impacts in gastrointestinal development including at early stages and is a valuable resource for further studies on the genetic and physiological control of the response of the animal to feed variations.


Animal Nutritional Physiological Phenomena , Breeding/standards , Chickens/physiology , Digestion/physiology , Gastrointestinal Tract/growth & development , Animal Feed/analysis , Animals , Breeding/methods , Chickens/blood , Chickens/genetics , Chickens/growth & development , Diet/veterinary , Female , Male
13.
PLoS One ; 16(1): e0245216, 2021.
Article En | MEDLINE | ID: mdl-33429419

Triploid, sterile Atlantic salmon (Salmo salar) could make a contribution to the development of the farming industry, but uncertainties about the performance and welfare of triploids have limited their adoption by farmers. In this study, we compared the ontogeny of digestive tract morphology and enzyme activities (pepsin, trypsin, chymotrypsin, alkaline phosphatase and aminopeptidase) of diploid and triploid Atlantic salmon. Fish were fed diets based on fishmeal (STD) or a mix of fishmeal and hydrolysed fish proteins (HFM) whilst being reared at low temperature from start-feeding to completion of the parr-smolt transformation. Fish weights for each ploidy and feed combination were used to calculate thermal growth coefficients (TGCs) that spanned this developmental period, and the data were used to examine possible relationships between enzyme activities and growth. At the end of the experiment, faeces were collected and analyzed to determine the apparent digestibility coefficients (ADCs) of the dietary amino acids (AAs). Digestive tract histo-morphology did not differ substantially between ploidies and generally reflected organ maturation and functionality. There were no consistent differences in proteolytic enzyme activities resulting from the inclusion of HFM in the diet, nor was there improved digestibility and AA bioavailability of the HFM feed in either diploid or triploid fish. The triploid salmon had lower ADCs than diploids for most essential and non-essential AAs in both diets (STD and HFM), but without there being any indication of lower intestinal protease activity in triploid fish. When trypsin-to-chymotrypsin activity and trypsin and alkaline phosphatase (ALP) ratios (T:C and T:ALP, respectively) were considered in combination with growth data (TGC) low T:C and T:ALP values coincided with times of reduced fish growth, and vice versa, suggesting that T:C and T:ALP may be used to predict recent growth history and possible growth potential.


Animal Feed , Diploidy , Fish Proteins , Gastrointestinal Tract , Protein Hydrolysates/pharmacology , Salmo salar , Triploidy , Animals , Fish Proteins/genetics , Fish Proteins/metabolism , Fish Proteins/pharmacology , Gastrointestinal Tract/anatomy & histology , Gastrointestinal Tract/enzymology , Gastrointestinal Tract/growth & development , Salmo salar/anatomy & histology , Salmo salar/genetics , Salmo salar/growth & development
14.
Lett Appl Microbiol ; 73(1): 20-25, 2021 Jul.
Article En | MEDLINE | ID: mdl-33386625

Sialic acid (N-acetylneuraminic acid), a 9-carbon monosaccharide, has been widely studied in immunology, oncology and neurology. However, the effects of sialic acid on organ and intestinal development, liver function and gut microbiota were rarely studied. In this study, we found that oral sialic acid tended to increase the relative weight of liver and decreased the serum aspartate aminotransferase (GPT) activity. In addition, sialic acid treatment markedly reduced gut villus length, depth, the ratio of villus length/depth (L/D), areas, width and the number of goblet cells. Furthermore, gut microbes were changed in response to oral sialic acid, such as Staphylococcus lentus, Corynebacterium stationis, Corynebacterium urealyticum, Jeotgalibaca sp_PTS2502, Ignatzschineria indica, Sporosarcina pasteurii, Sporosarcina sp_HW10C2, Facklamia tabacinasalis, Oblitimonas alkaliphila, Erysipelatoclostridium ramosum, Blautia sp_YL58, Bacteroids thetaiotaomicron, Morganella morganii, Clostridioides difficile, Helicobacter tryphlonius, Clostridium sp_Clone47, Alistipes finegoldii, [pseudomonas]_geniculata and Pseudomonas parafulva at the species level. In conclusion, oral sialic acid altered the intestinal pathological state and microbial compositions, and the effect of sialic acid on host health should be further studied.


Biodiversity , Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/drug effects , Liver/drug effects , N-Acetylneuraminic Acid/pharmacology , Administration, Oral , Animals , Aspartate Aminotransferases/blood , Enzyme Activation/drug effects , Gastrointestinal Tract/growth & development , Mice , N-Acetylneuraminic Acid/administration & dosage
15.
Int J Mol Sci ; 22(2)2021 Jan 18.
Article En | MEDLINE | ID: mdl-33477687

Parasympathetic signalling via muscarinic acetylcholine receptors (mAChRs) regulates gastrointestinal smooth muscle function. In most instances, the mAChR population in smooth muscle consists mainly of M2 and M3 subtypes in a roughly 80% to 20% mixture. Stimulation of these mAChRs triggers a complex array of biochemical and electrical events in the cell via associated G proteins, leading to smooth muscle contraction and facilitating gastrointestinal motility. Major signalling events induced by mAChRs include adenylyl cyclase inhibition, phosphoinositide hydrolysis, intracellular Ca2+ mobilisation, myofilament Ca2+ sensitisation, generation of non-selective cationic and chloride currents, K+ current modulation, inhibition or potentiation of voltage-dependent Ca2+ currents and membrane depolarisation. A lack of ligands with a high degree of receptor subtype selectivity and the frequent contribution of multiple receptor subtypes to responses in the same cell type have hampered studies on the signal transduction mechanisms and functions of individual mAChR subtypes. Therefore, novel strategies such as genetic manipulation are required to elucidate both the contributions of specific AChR subtypes to smooth muscle function and the underlying molecular mechanisms. In this article, we review recent studies on muscarinic function in gastrointestinal smooth muscle using mAChR subtype-knockout mice.


Gastrointestinal Tract/metabolism , Muscle, Smooth/metabolism , Receptor, Muscarinic M2/genetics , Receptor, Muscarinic M3/genetics , Animals , GTP-Binding Proteins/genetics , Gastrointestinal Tract/growth & development , Gastrointestinal Tract/pathology , Mice, Knockout/genetics , Muscle Contraction/genetics , Muscle, Smooth/growth & development , Signal Transduction/genetics
16.
Nutrients ; 14(1)2021 Dec 31.
Article En | MEDLINE | ID: mdl-35011069

BACKGROUND: Delayed onset of minimal enteral nutrition compromises the immune response of preterm infants, increasing the risk of colonization and clinical complications (e.g., late-onset sepsis). This study aimed to analyze associations between late-onset sepsis in very low birth weight infants (<1500 g) and days of parenteral nutrition, days to reach full enteral nutrition, and maternal and nutritional factors. METHODS: A cross-sectional study was carried out with very low birth weight infants admitted to a neonatal intensive care unit (NICU) of a reference maternity hospital of high-risk deliveries. Data regarding days of parenteral nutrition, days to reach full enteral nutrition, fasting days, extrauterine growth restriction, and NICU length of stay were extracted from online medical records. Late-onset sepsis was diagnosed (clinical or laboratory) after 48 h of life. Chi-squared, Mann-Whitney tests, and binary logistic regression were applied. RESULTS: A total of 97 preterm infants were included. Of those, 75 presented late-onset sepsis with clinical (n = 40) or laboratory (n = 35) diagnosis. Maternal urinary tract infection, prolonged parenteral nutrition (>14 days), and extrauterine growth restriction presented 4.24-fold, 4.86-fold, and 4.90-fold higher chance of late-onset sepsis, respectively. CONCLUSION: Very low birth weight infants with late-onset sepsis had prolonged parenteral nutrition and took longer to reach full enteral nutrition. They also presented a higher prevalence of extrauterine growth restriction than infants without late-onset sepsis.


Infant Nutritional Physiological Phenomena/physiology , Infant, Premature/physiology , Infant, Very Low Birth Weight/physiology , Neonatal Sepsis/epidemiology , Neonatal Sepsis/physiopathology , Birth Weight , Cross-Sectional Studies , Enteral Nutrition/methods , Gastrointestinal Tract/growth & development , Gestational Age , Humans , Infant, Newborn , Infant, Very Low Birth Weight/immunology , Intensive Care, Neonatal/methods , Parenteral Nutrition/adverse effects , Time Factors
17.
Primates ; 62(2): 431-441, 2021 Mar.
Article En | MEDLINE | ID: mdl-33180215

Digestive tract measurements are often considered species specific, but little information exists on the degree to which they change during ontogeny within a species. Additionally, access to anatomical material from nondomestic species is often limited, with fixed tissues possibly representing the only available source, though the degree to which this material is representative in terms of dimensions and weight is debatable. In the present study, the macroscopic anatomy of the digestive tract (length of intestinal sections, and tissue weights of stomach and intestines) of 58 Lemur catta [ranging in age from 1 month (neonates) to 25 years], which had been stored frozen (n = 27) or fixed in formalin (n = 31), was quantified. Particular attention was paid to the caecum and the possible presence of an appendix. The intraspecific allometric scaling of body mass (BM)0.46[0.40;0.51] for total intestine length and BM0.48[0.41;0.54] for small intestine length was higher than the expected geometric scaling of BM0.33, and similar to that reported in the literature for interspecific scaling. This difference in scaling is usually explained by the hypothesis that, to maintain optimal absorption, the diameter of the intestinal tube cannot increase geometrically. Therefore, geometric volume gain of increasing body mass is accommodated for by more-than-geometric length scaling. According to the literature, not all L. catta have an appendix. No appendix was found in the specimens in the present study. The proportions of length measurements did not change markedly during ontogeny, indicating that the proportions of the foetus are representative of those of the adult animal. By contrast, width and tissue-mass scaling of the caecum indicated disproportionate growth of this organ during ontogeny that was not reflected in its length. Compared to overall intraspecific variation, the method of storage (frozen vs. formalin) had no relevant impact on length or weight measurements.


Gastrointestinal Tract/anatomy & histology , Lemur/anatomy & histology , Specimen Handling/methods , Animals , Appendix , Body Weight , Cecum/anatomy & histology , Formaldehyde , Freezing , Gastrointestinal Tract/growth & development , Lemur/growth & development
18.
Toxins (Basel) ; 12(10)2020 10 01.
Article En | MEDLINE | ID: mdl-33019707

The chronic intake of naturally multi-mycotoxin contaminated feed by broilers with or without titers of Yeast Cell Wall Extract (YCWE, a.k.a Mycosorb A+®), was investigated. Day-old male Cobb chicks (1600 birds, 64 pens, 25 birds/pen) were randomly allocated to diets of control (CON); diet containing mycotoxins (MT); CON + 0.2% YCWE; MT + 0.025% YCWE; MT + 0.05% YCWE; MT + 0.1% YCWE; MT + 0.2% YCWE; and MT + 0.4% YCWE. Growth performance, blood biochemical parameters and gut health were recorded over 42 days. Compared with CON, MT had reduced body weight (BW) and increased feed conversion ratio (FCR) on days 35 and 42 with increased duodenal crypt depth and fewer goblet cells. Furthermore, European Poultry Production Efficiency (EPEF) was reduced for MT versus CON. Feeding MT + 0.2% YCWE improved BW, lowered FCR, reduced crypt depth, increased goblet cell count and improved EPEF. Considering titration of YCWE (0 to 0.4%) during mycotoxin challenge, a cubic effect was observed for FCR with NC + 0.2% YCWE having the lowest FCR. These findings suggest that chronic consumption of multiple Fusarium mycotoxins present in common field concentrations can negatively impact broiler performance and gut health while inclusion of YCWE, particularly 0.2%, could be effective in counteracting mycotoxins.


Animal Feed/microbiology , Cell Wall/metabolism , Chickens/growth & development , Dietary Supplements , Food Microbiology , Fusarium/metabolism , Mycotoxins/toxicity , Yeasts/metabolism , Age Factors , Animals , Animals, Newborn , Chickens/metabolism , Gastrointestinal Tract/growth & development , Male , Mycotoxins/metabolism , Weight Gain
19.
Gene Expr Patterns ; 38: 119149, 2020 12.
Article En | MEDLINE | ID: mdl-33007443

The Atlantic salmon has been studied extensively, particularly as a model for understanding the genetic and environmental contributions to the evolution and development of life history traits. Expression pattern analysis in situ, however, is mostly lacking in salmon. We examine the embryonic developmental expression of six6, a candidate gene previously identified to be associated with spawning ecotypes and age at sexual maturity, in Atlantic salmon. Six6 is a member of the sine oculis homeobox family of transcription factors and is known to regulate eye and brain development in other vertebrates. We assay the expression of this gene in embryonic Atlantic salmon Salmo salar by whole-mount in situ hybridization. In line with earlier studies in other vertebrate species, we find conserved expression in the developing brain and sensory organs, including optic and olfactory primordia. However, we also find previously unreported domains of expression that suggest additional roles in axial and appendicular development, cardiovascular, intestinal, and sensory organogenesis. Each of these systems are important in the sensory ecology of Atlantic salmon, suggesting it is plausible that six6 may have pleiotropic roles in this complex phenotype.


Fish Proteins/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Salmo salar/genetics , Animals , Brain/growth & development , Brain/metabolism , Cardiovascular System/growth & development , Cardiovascular System/metabolism , Fish Proteins/metabolism , Gastrointestinal Tract/growth & development , Gastrointestinal Tract/metabolism , Homeodomain Proteins/metabolism , Salmo salar/growth & development
20.
Arch Anim Nutr ; 74(6): 496-511, 2020 Dec.
Article En | MEDLINE | ID: mdl-32967440

The use of natural antioxidants, in particular polyphenols such as dihydroquercetin (DHQ), in animal nutrition has recently increased in popularity. This may partly be due to the risk of increased incidences of heat stress associated with raising livestock in warmer ambient temperatures, facilitated by global warming, reducing antioxidant capacity. The current research demonstrates the effect of dietary DHQ, vitaminEand standard or high ambient temperatures on growth performance, energy and nutrient metabolism, gastrointestinal tract (GIT) development, jejunal villus morphometry and antioxidant status in broiler chickens. Each of the four experimental diets was fed to 16 pens of five birds, which were allocated to four rooms (four pens in each room). The temperature in two rooms was maintained at aconstant 35°C (high temperature; HT), and the temperature in the other two rooms was gradually reduced from 27°C at 7 dof age to 22°C at 20 dof age (standard temperature; ST). Rearing birds at HT reduced feed intake, weight gain, weight of small intestine, total GIT, liver, spleen, heart, villus height, villus surface area and lowered blood glutationperoxidase (GSH-Px). Dietary DHQ increased blood GSH-Px and total antioxidant status, increased heart weight and reduced caecal size. When fed separately, DHQ and vitamin E improved hepatic vitamin E concentration. Feeding vitamin Eincreased spleen and liver weights. When fed together, DHQ and vitamin Ereduced villus height, villus height to crypt depth ratio and villus surface area. Temperature and antioxidants did not affect energy and nutrient metabolism. There were no effects of dietary antioxidants on growth performance of broiler chickens and there were no mortalities. At present, it is unclear if feeding antioxidants (in particular DHQ) at different levels, using different dietary formulations, and rearing birds under arange of environmental conditions may be effective at enhancing production performance and bird health in hot ambient climates.


Antioxidants/metabolism , Chickens/metabolism , Gastrointestinal Tract/growth & development , Jejunum/anatomy & histology , Quercetin/analogs & derivatives , Vitamin E/metabolism , Vitamins/metabolism , Animal Feed/analysis , Animals , Antioxidants/administration & dosage , Chickens/growth & development , Diet/veterinary , Dietary Supplements/analysis , Dose-Response Relationship, Drug , Gastrointestinal Tract/drug effects , Jejunum/drug effects , Male , Organ Size/drug effects , Quercetin/administration & dosage , Quercetin/metabolism , Random Allocation , Temperature , Vitamin E/administration & dosage , Vitamins/administration & dosage
...