Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.220
Filter
1.
Science ; 382(6670): 527-528, 2023 11 03.
Article in English | MEDLINE | ID: mdl-37917691

ABSTRACT

Glial cells in the gut are specialized to fine-tune intestinal function.


Subject(s)
Intestines , Neuroglia , Neuroglia/physiology , Intestines/innervation , Intestines/physiology , Intestines/ultrastructure , Animals , Mice , Myenteric Plexus/physiology , Myenteric Plexus/ultrastructure
2.
Cell Rep ; 38(7): 110379, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35172130

ABSTRACT

Pluripotent-stem-cell-derived human intestinal organoids (HIOs) model some aspects of intestinal development and disease, but current culture methods do not fully recapitulate the diverse cell types and complex organization of the human intestine and are reliant on 3D extracellular matrix or hydrogel systems, which limit experimental control and translational potential for regenerative medicine. We describe suspension culture as a simple, low-maintenance method for culturing HIOs and for promoting in vitro differentiation of an organized serosal mesothelial layer that is similar to primary human intestinal serosal mesothelium based on single-cell RNA sequencing and histological analysis. Functionally, HIO serosal mesothelium has the capacity to differentiate into smooth-muscle-like cells and exhibits fibrinolytic activity. An inhibitor screen identifies Hedgehog and WNT signaling as regulators of human serosal mesothelial differentiation. Collectively, suspension HIOs represent a three-dimensional model to study the human serosal mesothelium.


Subject(s)
Epithelium/growth & development , Intestines/growth & development , Organoids/growth & development , Serous Membrane/growth & development , Tissue Culture Techniques , Alginates/pharmacology , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Line , Collagen/pharmacology , Drug Combinations , Epithelium/drug effects , Hedgehog Proteins/metabolism , Humans , Intestines/ultrastructure , Laminin/pharmacology , Muscle, Smooth/cytology , Organoids/drug effects , Organoids/ultrastructure , Proteoglycans/pharmacology , Serous Membrane/drug effects , Serous Membrane/ultrastructure , Signal Transduction/drug effects , Suspensions , Wnt Proteins/metabolism
3.
Article in English | MEDLINE | ID: mdl-34562624

ABSTRACT

Ambush-foraging snakes that ingest large meals might undergo several months without eating when they use the internal reserves to support the energetic costs of living. Then, morphological and physiological processes might be orchestrated during the transition from fasting to the postprandial period to rapidly use the energetic stores while the metabolic rate is elevated in response to food intake. To understand the patterns of substrates deposition after feeding, we accessed the morphological and biochemical response in Boa constrictor snakes after two months of fasting and six days after feeding. We followed the plasma levels of glucose, total proteins, and total lipids, and we performed the stereological ultrastructural analysis of the liver and the proximal region of the intestine to quantify glycogen granules and lipid droplets. In the same tissues and stomach, we measured the activity of the enzyme fructose-1,6-biphosphatase (FBPase1) involved in the gluconeogenic pathway, and we measured pyruvate kinase (PK) and lactate dehydrogenase (LDH) enzymatic activities involved in the anaerobic pathway in the liver. Briefly, our results indicated an increase in boas' plasma glucose one day after meal intake compared to unfed snakes. The hepatic glycogen reserves were continuously restored within days after feeding. Also, the enzymes involved in the energetic pathways increased activity six days after feeding in the liver. These findings suggest a quick restoring pattern of energetic stores during the postprandial period.


Subject(s)
Boidae/physiology , Animals , Blood Glucose/metabolism , Blood Proteins/metabolism , Boidae/blood , Eating/physiology , Energy Metabolism , Feeding Behavior/physiology , Gluconeogenesis , Homeostasis , Intestines/metabolism , Intestines/ultrastructure , Lipids/blood , Liver/metabolism , Liver/ultrastructure , Liver Glycogen/metabolism , Microscopy, Electron, Transmission , Postprandial Period/physiology
4.
Nutrients ; 13(8)2021 Jul 23.
Article in English | MEDLINE | ID: mdl-34444677

ABSTRACT

Caffeine, a methylxanthine derived from plants, is the most widely consumed ingredient in daily life. Therefore, it is necessary to investigate the effects of caffeine intake on essential biological activities. In this study, we attempted to determine the possible anti-aging effects of long-term caffeine intake in the intestine of an aged Caenorhabditis elegans model. We examined changes in intestinal integrity, production of vitellogenin (VIT), and mitochondrial function after caffeine intake. To evaluate intestinal aging, actin-5 (ACT-5) mislocalization, lumenal expansion, and intestinal colonization were examined after caffeine intake, and the levels of vitellogenesis as well as the mitochondrial activity were measured. We found that the long-term caffeine intake (10 mM) in the L4-stage worms at 25 °C for 3 days suppressed ACT-5 mislocalization. Furthermore, the level of autophagy, which is normally increased in aging animals, was significantly reduced in these animals, and their mitochondrial functions improved after caffeine intake. In addition, the caffeine-ingesting aging animals showed high resistance to oxidative stress and increased the expression of antioxidant proteins. Taken together, these findings reveal that caffeine may be a potential anti-aging agent that can suppress intestinal atrophy during the progression of intestinal aging.


Subject(s)
Aging/physiology , Caenorhabditis elegans/physiology , Caffeine/administration & dosage , Intestines/physiology , Mitochondria/physiology , Vitellogenesis/drug effects , Actins/analysis , Animals , Animals, Genetically Modified , Caenorhabditis elegans/drug effects , Intestines/ultrastructure , Mitochondria/drug effects , Models, Animal , Oxidative Stress/drug effects
5.
Int J Biol Sci ; 17(7): 1693-1707, 2021.
Article in English | MEDLINE | ID: mdl-33994854

ABSTRACT

This study is to investigate the relationship between berberine (BBR) and mitochondrial complex I in lipid metabolism. BBR reversed high-fat diet-induced obesity, hepatic steatosis, hyperlipidemia and insulin resistance in mice. Fatty acid consumption, ß-oxidation and lipogenesis were attenuated in liver after BBR treatment which may be through reduction in SCD1, FABP1, CD36 and CPT1A. BBR promoted fecal lipid excretion, which may result from the reduction in intestinal CD36 and SCD1. Moreover, BBR inhibited mitochondrial complex I-dependent oxygen consumption and ATP synthesis of liver and gut, but no impact on activities of complex II, III and IV. BBR ameliorated mitochondrial swelling, facilitated mitochondrial fusion, and reduced mtDNA and citrate synthase activity. BBR decreased the abundance and diversity of gut microbiome. However, no change in metabolism of recipient mice was observed after fecal microbiota transplantation from BBR treated mice. In primary hepatocytes, BBR and AMPK activator A769662 normalized oleic acid-induced lipid deposition. Although both the agents activated AMPK, BBR decreased oxygen consumption whereas A769662 increased it. Collectively, these findings indicated that BBR repressed complex I in gut and liver and consequently inhibited lipid metabolism which led to alleviation of obesity and fatty liver. This process was independent of intestinal bacteria.


Subject(s)
Berberine/pharmacology , Electron Transport Complex I/deficiency , Intestines/metabolism , Lipid Metabolism Disorders/drug therapy , Lipid Metabolism/drug effects , Liver/metabolism , Animals , Disease Models, Animal , Electron Transport Complex I/drug effects , Electron Transport Complex I/metabolism , Intestines/drug effects , Intestines/ultrastructure , Lipid Metabolism Disorders/metabolism , Lipid Metabolism Disorders/pathology , Liver/drug effects , Liver/ultrastructure , Male , Mice , Mice, Inbred AKR , Microscopy, Electron, Transmission , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Oxidation-Reduction
6.
Nat Commun ; 12(1): 2886, 2021 05 17.
Article in English | MEDLINE | ID: mdl-34001900

ABSTRACT

The brush border is comprised of microvilli surface protrusions on the apical surface of epithelia. This specialized structure greatly increases absorptive surface area and plays crucial roles in human health. However, transcriptional regulatory networks controlling brush border genes are not fully understood. Here, we identify that hepatocyte nuclear factor 4 (HNF4) transcription factor is a conserved and important regulator of brush border gene program in multiple organs, such as intestine, kidney and yolk sac. Compromised brush border gene signatures and impaired transport were observed in these tissues upon HNF4 loss. By ChIP-seq, we find HNF4 binds and activates brush border genes in the intestine and kidney. H3K4me3 HiChIP-seq identifies that HNF4 loss results in impaired chromatin looping between enhancers and promoters at gene loci of brush border genes, and instead enhanced chromatin looping at gene loci of stress fiber genes in the intestine. This study provides comprehensive transcriptional regulatory mechanisms and a functional demonstration of a critical role for HNF4 in brush border gene regulation across multiple murine epithelial tissues.


Subject(s)
Gene Expression Regulation , Hepatocyte Nuclear Factor 4/genetics , Intestinal Mucosa/metabolism , Kidney/metabolism , Microvilli/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Yolk Sac/metabolism , Animals , Epithelium/metabolism , Gene Expression Profiling/methods , Hepatocyte Nuclear Factor 4/metabolism , Humans , Intestines/ultrastructure , Kidney/ultrastructure , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission , Receptors, Cytoplasmic and Nuclear/metabolism , Reverse Transcriptase Polymerase Chain Reaction
7.
Sci Rep ; 11(1): 3544, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33574405

ABSTRACT

Global anal cancer incidence is increasing. High resolution anoscopy (HRA) currently screens for anal cancer, although the definitive test remains unknown. To improve on intraluminal imaging of the anal canal, we conducted a first-in-human study to determine feasibility and safety of a high-resolution, wide field-of-view scanning endoscope. Fourteen patients, under an IRB-approved clinical study, underwent exam under anesthesia, HRA, and imaging with the experimental device. HRA findings were photographed using an in-line camera attached to the colposcope and compared with the scanning endoscope images. Patients were followed up within 2 weeks of the procedure. The imaging device is inserted into the anal canal and the intraluminal surface is digitally photographed in 10 s and uploaded to a computer monitor for review. Ten patients completed imaging with the device. Three patients were not imaged due to severe anal stenosis. One patient was not imaged due to technical device malfunction. The device images were compared to the HRA images. No adverse event attributable to the device was reported. The intraluminal scanning endoscope can be used for circumferential anal canal imaging and is safe for clinical use. Future clinical studies are needed to evaluate the performance of this device.


Subject(s)
Anus Diseases/diagnosis , Early Detection of Cancer , Endoscopes, Gastrointestinal , Intestines/diagnostic imaging , Precancerous Conditions/diagnosis , Aged , Anal Canal/diagnostic imaging , Anal Canal/pathology , Anus Diseases/diagnostic imaging , Anus Diseases/pathology , Colposcopes/standards , Diagnostic Imaging/instrumentation , Diagnostic Imaging/methods , Feasibility Studies , Female , Humans , Intestines/ultrastructure , Male , Middle Aged , Precancerous Conditions/diagnostic imaging , Precancerous Conditions/pathology
8.
Nat Commun ; 12(1): 56, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33397922

ABSTRACT

RAC1 activity is critical for intestinal homeostasis, and is required for hyperproliferation driven by loss of the tumour suppressor gene Apc in the murine intestine. To avoid the impact of direct targeting upon homeostasis, we reasoned that indirect targeting of RAC1 via RAC-GEFs might be effective. Transcriptional profiling of Apc deficient intestinal tissue identified Vav3 and Tiam1 as key targets. Deletion of these indicated that while TIAM1 deficiency could suppress Apc-driven hyperproliferation, it had no impact upon tumourigenesis, while VAV3 deficiency had no effect. Intriguingly, deletion of either gene resulted in upregulation of Vav2, with subsequent targeting of all three (Vav2-/- Vav3-/- Tiam1-/-), profoundly suppressing hyperproliferation, tumourigenesis and RAC1 activity, without impacting normal homeostasis. Critically, the observed RAC-GEF dependency was negated by oncogenic KRAS mutation. Together, these data demonstrate that while targeting RAC-GEF molecules may have therapeutic impact at early stages, this benefit may be lost in late stage disease.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Guanine Nucleotide Exchange Factors/metabolism , Intestines/pathology , Signal Transduction , rac1 GTP-Binding Protein/metabolism , Adenomatous Polyposis Coli Protein/metabolism , Animals , Carcinogenesis/genetics , Homeostasis , Intestines/ultrastructure , Mice, Knockout , Mutation/genetics , Organ Specificity , Phenotype , Proto-Oncogene Proteins c-vav/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , T-Lymphoma Invasion and Metastasis-inducing Protein 1/metabolism , Up-Regulation , Wnt Signaling Pathway
9.
Int J Mol Sci ; 21(21)2020 11 03.
Article in English | MEDLINE | ID: mdl-33153048

ABSTRACT

The intestine is an organ essential to organismal nutrient absorption, metabolic control, barrier function and immunoprotection. The Caenorhabditis elegans intestine consists of 20 cells harboring a dense intermediate filament network positioned below the apical plasma membrane that forms a junction-anchored sheath around the intestinal lumen. This evolutionarily conserved arrangement provides mechanical and overall stress-protection, and it serves as an important model for deciphering the role of intestinal architecture in metazoan biology. We recently reported that the loss-of-function mutation of the intestinal intermediate filament organizer IFO-1 perturbs this architecture, leading to reduced body size and reproduction. Here, we demonstrate that the IFO-1 mutation dramatically affects cholesterol metabolism. Mutants showed an increased sensitivity to cholesterol depletion, reduced cholesterol uptake, and cholesterol transfer to the gonads, which is also observed in worms completely lacking an intermediate filament network. Accordingly, we found striking similarities to transcriptome and lipidome profiles of a nuclear hormone receptor (NHR)-8 mutant. NHR-8 is homologous to mammalian LXR (liver X receptor) that serves as a sterol sensor and transcriptional regulator of lipid metabolism. Remarkably, increasing exogenous cholesterol partially rescues the developmental retardation in IFO-1 mutants. Our results uncover a novel link of the intestinal intermediate filament cytoskeleton to cholesterol metabolism that contributes to compromised growth and reproduction.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans , Cholesterol/metabolism , Intermediate Filament Proteins/genetics , Lipid Metabolism/genetics , Animals , Animals, Genetically Modified , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Cholesterol/pharmacology , Embryo, Nonmammalian , Gene Expression Regulation, Developmental/drug effects , Intermediate Filament Proteins/metabolism , Intermediate Filaments/metabolism , Intestinal Mucosa/embryology , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Intestines/embryology , Intestines/physiology , Intestines/ultrastructure , Lipid Metabolism/drug effects , Lipidomics , Receptors, Cytoplasmic and Nuclear/physiology , Transcriptome/drug effects
10.
Sci Rep ; 10(1): 11507, 2020 07 13.
Article in English | MEDLINE | ID: mdl-32661347

ABSTRACT

Gut hyperpermeability can be caused by either apoptosis of the intestinal epithelium or altered status, permeability or porosity of tight junctions. This project aims to elucidate these mechanisms in the early phase of sepsis. Eighteen male wild type mice were randomized to two groups. All mice received one single gavage of fluorescein isothiocyanate (FITC) dextran 30 min before intervention. One group (n = 10) underwent cecal ligation and puncture to induce sepsis. The other group (n = 8) was sham operated. Septic animals exhibited significantly increased permeability for FITC 8 h post-operatively. Significantly increased serum interleukin-6, tumor-necrosis-factor-alpha and interleukin-1-beta confirmed sepsis. Septic animals showed significant bowel wall inflammation of ileum and colon samples. PCR revealed significantly increased expression of claudin-2 and decreased expressions of claudin-4, tight-junction-protein-1 and occludin-1 resembling increased permeability of tight junctions. However, these alterations could not be confirmed at the protein level. Light microscopy revealed significant dilatation of intercellular spaces at the basal sections of intestinal epithelial cells (IEC) in septic animals confirmed by increased intercellular spaces at the level of tight junctions and adherens junctions in electron microscopy (TEM). In small angle X-ray scattering no increase in number or size of nanopores could be shown in the bowel wall. HOECHST staining and PCR of ileum samples for apoptosis markers proofed no relevant differences in intestinal epithelial cell apoptosis between the groups. Intestinal hyperpermeability in septic animals was most likely caused by alterations of the intercellular contacts and not by apoptosis or increased size/number of nanopores of intestinal epithelial cells in this murine model of early sepsis.


Subject(s)
Epithelial Cells/ultrastructure , Intestines/ultrastructure , Sepsis/pathology , Tight Junctions/ultrastructure , Animals , Apoptosis/genetics , Cecum/pathology , Cecum/ultrastructure , Colon/pathology , Colon/ultrastructure , Disease Models, Animal , Epithelial Cells/pathology , Humans , Ileum/pathology , Ileum/ultrastructure , Intestinal Mucosa/pathology , Intestinal Mucosa/ultrastructure , Intestines/pathology , Mice , Permeability , Sepsis/metabolism , Tight Junctions/pathology
11.
J Endocrinol ; 247(1): 11-24, 2020 10.
Article in English | MEDLINE | ID: mdl-32698143

ABSTRACT

Fructose dietary intake affects the composition of the intestinal microbiota and influences the development of hepatic steatosis. Endotoxins produced by gram-negative bacteria alter intestinal permeability and cause bacterial translocation. This study evaluated the effects of gut microbiota modulation by a purified PPAR-alpha agonist (WY14643), a DPP-4 inhibitor (linagliptin), or their association on intestinal barrier integrity, endotoxemia, and hepatic energy metabolism in high-fructose-fed C57BL/6 mice. Fifty mice were divided to receive the control diet (C group) or the high-fructose diet (HFRU) for 12 weeks. Subsequently, the HFRU group was divided to initiate the treatment with PPAR-alpha agonist (3.5 mg/kg/BM) and DPP-4 inhibitor (15 mg/kg/BM). The HFRU group had glucose intolerance, endotoxemia, and dysbiosis (with increased Proteobacteria) without changes in body mass in comparison with the C group. HFRU group showed damaged intestinal ultrastructure, which led to liver inflammation and marked hepatic steatosis in the HFRU group when compared to the C group. PPAR-alpha activation and DPP-4 inhibition countered glucose intolerance, endotoxemia, and dysbiosis, ameliorating the ultrastructure of the intestinal barrier and reducing Tlr4 expression in the liver of treated animals. These beneficial effects suppressed lipogenesis and mitigated hepatic steatosis. In conclusion, the results herein propose a role for PPAR-alpha activation, DPP-4 inhibition, and their association in attenuating hepatic steatosis by gut-liver axis modulation in high-fructose mice model. These observations suggest these treatments as potential targets to treat hepatic steatosis and avoid its progression.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Fructose/administration & dosage , Gastrointestinal Microbiome/drug effects , Linagliptin/pharmacology , Liver/drug effects , PPAR alpha/physiology , Animals , Blood Glucose/analysis , Diet , Endotoxemia/prevention & control , Fatty Liver/prevention & control , Gastrointestinal Microbiome/physiology , Intestines/drug effects , Intestines/ultrastructure , Lipogenesis/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , PPAR alpha/drug effects , Peroxisome Proliferators , Pyrimidines/pharmacology
12.
Immunity ; 52(3): 557-570.e6, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32160523

ABSTRACT

The intestine contains some of the most diverse and complex immune compartments in the body. Here we describe a method for isolating human gut-associated lymphoid tissues (GALTs) that allows unprecedented profiling of the adaptive immune system in submucosal and mucosal isolated lymphoid follicles (SM-ILFs and M-ILFs, respectively) as well as in GALT-free intestinal lamina propria (LP). SM-ILF and M-ILF showed distinct patterns of distribution along the length of the intestine, were linked to the systemic circulation through MAdCAM-1+ high endothelial venules and efferent lymphatics, and had immune profiles consistent with immune-inductive sites. IgA sequencing analysis indicated that human ILFs are sites where intestinal adaptive immune responses are initiated in an anatomically restricted manner. Our findings position ILFs as key inductive hubs for regional immunity in the human intestine, and the methods presented will allow future assessment of these compartments in health and disease.


Subject(s)
Adaptive Immunity/immunology , Immunity, Mucosal/immunology , Intestinal Mucosa/immunology , Intestines/immunology , Lymphoid Tissue/immunology , Adaptive Immunity/genetics , Animals , Flow Cytometry , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastric Mucosa/ultrastructure , Humans , Immunity, Mucosal/genetics , Immunoglobulin A/genetics , Immunoglobulin A/immunology , Immunoglobulin M/genetics , Immunoglobulin M/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Intestines/ultrastructure , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphoid Tissue/metabolism , Lymphoid Tissue/ultrastructure , Microscopy, Confocal , Microscopy, Electron, Scanning , Peyer's Patches/immunology , Peyer's Patches/metabolism , Peyer's Patches/ultrastructure , Sequence Analysis, DNA
13.
Ecotoxicol Environ Saf ; 193: 110372, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32114238

ABSTRACT

The objective of this study was to investigate the effects of fluorine at levels of 31, 431, 1237 mg/kg feed on cecum microbe, short-chain fatty acids (SCFAs) and intestinal barrier function of laying hens. The results showed that the intestinal morphology and ultrastructure were damaged by dietary high F intake. The mRNA expression levels of zonula occludens-1, zonula occludens-2, claudin-1, and claudin-4 were decreased in jejunum and ileum. However, the concentrations of serum diamine oxidase, and D-lactic acid and intestinal contents of interleukin 1 beta, interleukin 6, and Tumor necrosis factor-alpha were increased. Consistent with this, dietary high F intake altered the cecum microbiota, with increasing the concentration of pathogens, such as Proteobacteria and Escherichia-Shigella, as well as, decreasing the contents of beneficial bacteria, such as Lactobacillus, and expectedly, reduced the SCFAs concentrations. In conclusion, the actual results confirmed that (1) high dietary F intake could damage the intestinal structure and function, with impaired intestinal barrier and intestinal inflammation, and (2) destroy the cecum microbial homeostasis, and decrease the concentrations of SCFAs, which aggravate the incidence of intestinal inflammation in laying hens.


Subject(s)
Cecum/microbiology , Chickens , Fluorides/toxicity , Intestines/drug effects , Microbiota/drug effects , Animals , Chickens/anatomy & histology , Chickens/metabolism , Claudins/metabolism , Cytokines/metabolism , Diet , Fatty Acids, Volatile/metabolism , Female , Intestines/pathology , Intestines/ultrastructure , Lactobacillus , Tight Junctions/metabolism
14.
Sci Rep ; 10(1): 3142, 2020 02 21.
Article in English | MEDLINE | ID: mdl-32081918

ABSTRACT

The abundance and diversity of intermediate filaments (IFs) in the C. elegans intestine indicate important contributions to intestinal function and organismal wellbeing. Fluorescent IF reporters localize below the actin-rich brush border and are highly enriched in the lumen-enveloping endotube, which is attached to the C. elegans apical junction. Mapping intestinal viscoelasticity by contact-free Brillouin microscopy reveals that the IF-rich endotube is positioned at the interface between the stiff brush border and soft cytoplasm suggesting a mechanical buffering function to deal with the frequent luminal distortions occurring during food intake and movement. In accordance, depletion of IFB-2, IFC-2 and IFD-2 leads to intestinal lumen dilation although depletion of IFC-1, IFD-1 and IFP-1 do not. Ultrastructural analyses of loss of function mutants further show that IFC-2 mutants have a rarefied endotube and IFB-2 mutants lack an endotube altogether. Remarkably, almost all IFB-2- and IFC-2-deficient animals develop to fertile adults. But developmental retardation, reduced brood size, altered survival and increased sensitivity to microbial toxin, osmotic and oxidative stress are seen in both mutants albeit to different degrees. Taken together, we propose that individual intestinal IF polypeptides contribute in different ways to endotube morphogenesis and cooperate to cope with changing environments.


Subject(s)
Intermediate Filaments/metabolism , Intermediate Filaments/ultrastructure , Intestines/ultrastructure , Actin Cytoskeleton/metabolism , Actins/chemistry , Animals , Bacterial Proteins/metabolism , Caenorhabditis elegans , Caenorhabditis elegans Proteins/metabolism , Cytoskeleton/metabolism , Elasticity , Green Fluorescent Proteins/metabolism , Intermediate Filament Proteins/metabolism , Intestinal Mucosa/metabolism , Luminescent Proteins/metabolism , Microvilli/metabolism , Microvilli/ultrastructure , Mutation , Oxidative Stress , Viscosity
15.
Nat Med ; 26(4): 599-607, 2020 04.
Article in English | MEDLINE | ID: mdl-32094926

ABSTRACT

Mucosal immunity develops in the human fetal intestine by 11-14 weeks of gestation, yet whether viable microbes exist in utero and interact with the intestinal immune system is unknown. Bacteria-like morphology was identified in pockets of human fetal meconium at mid-gestation by scanning electron microscopy (n = 4), and a sparse bacterial signal was detected by 16S rRNA sequencing (n = 40 of 50) compared to environmental controls (n = 87). Eighteen taxa were enriched in fetal meconium, with Micrococcaceae (n = 9) and Lactobacillus (n = 6) the most abundant. Fetal intestines dominated by Micrococcaceae exhibited distinct patterns of T cell composition and epithelial transcription. Fetal Micrococcus luteus, isolated only in the presence of monocytes, grew on placental hormones, remained viable within antigen presenting cells, limited inflammation ex vivo and possessed genomic features linked with survival in the fetus. Thus, viable bacteria are highly limited in the fetal intestine at mid-gestation, although strains with immunomodulatory capacity are detected in subsets of specimens.


Subject(s)
Bacteria/growth & development , Fetus/microbiology , Gastrointestinal Microbiome , Intestines/microbiology , Microbial Viability , Autopsy , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacterial Typing Techniques , Female , Fetus/pathology , Fetus/ultrastructure , Gastrointestinal Microbiome/genetics , Gestational Age , Humans , Infant, Newborn , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Intestinal Mucosa/ultrastructure , Intestines/ultrastructure , Lactobacillus/classification , Lactobacillus/genetics , Lactobacillus/isolation & purification , Meconium/microbiology , Micrococcaceae/classification , Micrococcaceae/genetics , Micrococcaceae/isolation & purification , Pregnancy , Pregnancy Trimester, Second , RNA, Ribosomal, 16S/genetics
16.
Exp Mol Pathol ; 113: 104269, 2020 04.
Article in English | MEDLINE | ID: mdl-31202738

ABSTRACT

Supplemental oxygen is often used to treat neonates with respiratory disorders. Preclinical studies have demonstrated that neonatal hyperoxia injures the distal small intestine and activates nuclear factor-κB (NF-κB). Cathelicidin inhibits NF-κB activity and ameliorates lipopolysaccharide-induced intestinal barrier disruption in rats. Sprague-Dawley rat pups were reared in either room air (RA) or hyperoxia (85% O2) and were randomly treated with low-dose cathelicidin (4 mg/kg, LDC) and high-dose cathelicidin (HDC, 8 mg/kg) in 0.05 mL of normal saline (NS) administered intraperitoneally on postnatal days 1-6. The following six groups were obtained: RA + NS, RA + LDC, RA + HDC, O2 + NS, O2 + LDC, and O2 + HDC. The animals were sacrificed and the terminal ileum was removed for Western blot and histological analyses on postnatal day 7. The hyperoxia-reared rats exhibited significantly lower body weights, higher intestinal injury scores, lower occludin and ZO-1 expression, higher intestinal permeability and inducible IκB kinase inhibitor (IKKi) and NF-κB expression than the RA-reared rats. Cathelicidin treatment attenuated intestinal injury as evidenced by lower intestinal injury scores and intestinal permeability and higher intestinal barrier protein expression. The decrease in intestinal injury was accompanied by a decrease in IKKi and NF-κB. Cathelicidin attenuated hyperoxia-induced intestinal injury in the newborn rats, likely through NF-κB activity inhibition.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Hyperoxia/complications , Intestines/injuries , NF-kappa B/metabolism , Animals , Animals, Newborn , Body Weight/drug effects , Fatty Acid-Binding Proteins/metabolism , Female , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/ultrastructure , Intestines/drug effects , Intestines/pathology , Intestines/ultrastructure , Occludin/metabolism , Permeability , Rats, Sprague-Dawley , Zonula Occludens-1 Protein/metabolism , Cathelicidins
17.
Shock ; 54(1): 102-109, 2020 07.
Article in English | MEDLINE | ID: mdl-31361709

ABSTRACT

OBJECTIVE: Heatstroke can induce serious physiological dysfunction in the intestine. However, the underlying mechanisms of this condition are unknown, and therapeutic strategies are not available. In this study, we explored the role of endoplasmic reticulum (ER) stress signaling in this process and assessed whether pretreating mice with an inhibitor of ER stress could alleviate intestinal damage. METHODS: A heatstroke model was established in male mice. Mice were pretreated with 4-phenylbutyrate (4-PBA) before exposure to heat stress. Intestinal morphological changes were observed by hematoxylin and eosin (H&E) staining and transmission electron microscopy. The TUNEL assay was used to detect intestinal apoptosis. The expression of the ER stress-related proteins and apoptosis-related proteins was investigated by the Western blot assay. RESULTS: Compared with control group, mice with heatstroke exhibited evidence of intestinal injury and epithelial apoptosis, accompanied by significantly increased expression of ER stress-related proteins in the intestines. The intestinal injury score and level of intestinal epithelial apoptosis were significantly reduced after administration of 4-PBA. Furthermore, the levels of the intestinal ER stress-related proteins GRP78, PERK, p-eIF2α, ATF4, and CHOP were decreased after 4-PBA treatment. CONCLUSIONS: Our results indicate that the ER stress-mediated apoptosis pathway is activated during heat stress-induced intestinal injury. 4-PBA can inhibit heatstroke-induced intestinal ER stress and attenuate intestinal injury. We provide evidence that the beneficial effect of 4-PBA is closely related to the inhibition of ER stress-mediated apoptosis. These findings suggest that ER stress may be a novel therapeutic target in patients with heatstroke.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Heat Stroke/complications , Intestines/pathology , Phenylbutyrates/pharmacology , Animals , Endoplasmic Reticulum Chaperone BiP , Heat Stroke/pathology , In Situ Nick-End Labeling , Intestines/drug effects , Intestines/ultrastructure , Male , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission
18.
J Struct Biol ; 208(2): 174-181, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31476367

ABSTRACT

Assessing the internal morphology of Caenorhabditis elegans by a topographical technique like atomic force microscopy (AFM) is a challenging process. As a prerequisite for a successful image acquisition, direct contact between the structure of interest and the AFM probe needs to be established. To gain this insight into the morphology of cuticle and intestine in C. elegans before and after treatment with a tannin-enriched hydro-ethanolic extract from Combretum mucronatum, we developed an approach based on polyethylene glycol embedding, ultra-sectioning, de-embedding and hexamethyldisilazane-dehydration prior to measuring in ambient conditions by intermittent contact mode AFM. The used experimental protocol allowed a facile and fast insight into the ultrastructure of treated versus untreated C. elegans individuals, directly leading to the identification of treatment-associated morphological alterations in the cuticle but not the intestine of C. elegans. Additionally, the presented ultra-microtomy based protocol could allow future insight into virtually any tissue or organism by AFM.


Subject(s)
Caenorhabditis elegans/drug effects , Combretum/chemistry , Intestines/drug effects , Plant Extracts/pharmacology , Animals , Anthelmintics/chemistry , Anthelmintics/pharmacology , Caenorhabditis elegans/ultrastructure , Intestines/ultrastructure , Microscopy, Atomic Force , Plant Extracts/chemistry , Tannins/pharmacology
19.
Toxins (Basel) ; 11(6)2019 05 29.
Article in English | MEDLINE | ID: mdl-31146400

ABSTRACT

Tetrodotoxin (TTX) is an extremely toxic marine compound produced by different genera of bacteria that can reach humans through ingestion mainly of pufferfish but also of other contaminated fish species, marine gastropods or bivalves. TTX blocks voltage-gated sodium channels inhibiting neurotransmission, which in severe cases triggers cardiorespiratory failure. Although TTX has been responsible for many human intoxications limited toxicological data are available. The recent expansion of TTX from Asian to European waters and diversification of TTX-bearing organisms entail an emerging risk of food poisoning. This study is focused on the acute toxicity assessment of TTX administered to mice by oral gavage following macroscopic and microscopic studies. Necropsy revealed that TTX induced stomach swelling 2 h after administration, even though no ultrastructural alterations were further detected. However, transmission electron microscopy images showed an increase of lipid droplets in hepatocytes, swollen mitochondria in spleens, and alterations of rough endoplasmic reticulum in intestines as hallmarks of the cellular damage. These findings suggested that gastrointestinal effects should be considered when evaluating human TTX poisoning.


Subject(s)
Neurotoxins/toxicity , Tetrodotoxin/toxicity , Administration, Oral , Animals , Brain/drug effects , Brain/pathology , Brain/ultrastructure , Endoplasmic Reticulum, Rough/drug effects , Female , Intestines/drug effects , Intestines/pathology , Intestines/ultrastructure , Kidney/drug effects , Kidney/pathology , Kidney/ultrastructure , Liver/drug effects , Liver/pathology , Liver/ultrastructure , Lung/drug effects , Lung/pathology , Lung/ultrastructure , Mice , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/pathology , Myocardium/pathology , Myocardium/ultrastructure , Paralysis/chemically induced , Seizures/chemically induced , Spleen/drug effects , Spleen/pathology , Spleen/ultrastructure , Stomach/drug effects , Stomach/ultrastructure , Toxicity Tests, Acute
20.
Biofabrication ; 11(4): 045001, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31091514

ABSTRACT

A new in vitro gut microfluidic chip that mimics in vivo intestinal canal morphology and stimulation is developed to contribute to research into tissue engineering, and intestinal development and function. This strategy utilizes centrifugation to configure spatial cells along the side wall of a vertical cylinder-like microfluidic chamber, by which a tubular intestinal epithelium cell sheet is formed. Diverse intestinal cell lines are inoculated to address this approach. Furthermore, to generate microenvironmental stimulation, low-level centrifugation introduces fluid flow to this microfluidic system perpendicularly acting on cell sheet cultivation for several days. Fluid flow engenders the sectional cell sheet to bend toward the cell chamber lumen, which manifests an intestinal epithelium vaulted and wrinkle morphology. This may mimic the fluid flow existing in in vivo material transportation and the absorption of the gut epithelium barrier. In addition, the same fluid flow stimulation was reproduced in another Transwell system, which also exhibited a wrinkle epithelium cell sheet. Under fluid flow stimulation, some of the villus specific genes' expression level increased in the microfluidics and Transwell insert. Thus, this new centrifugation configuring gut microfluidic chip may offer novel insights into the research of intestinal structure and function.


Subject(s)
Intestines/physiology , Lab-On-A-Chip Devices , Tissue Engineering/methods , Animals , Cell Culture Techniques , Cell Line , Centrifugation , Equipment Design , Gene Expression Regulation , Humans , Intestinal Mucosa/physiology , Intestinal Mucosa/ultrastructure , Intestines/ultrastructure , Rats , Rheology
SELECTION OF CITATIONS
SEARCH DETAIL
...