Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 750
Filter
1.
EMBO J ; 43(16): 3466-3493, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38965418

ABSTRACT

The gut microbiota and their metabolites are closely linked to obesity-related diseases, such as type 2 diabetes, but their causal relationship and underlying mechanisms remain largely elusive. Here, we found that dysbiosis-induced tyramine (TA) suppresses high-fat diet (HFD)-mediated insulin resistance in both Drosophila and mice. In Drosophila, HFD increases cytosolic Ca2+ signaling in enterocytes, which, in turn, suppresses intestinal lipid levels. 16 S rRNA sequencing and metabolomics revealed that HFD leads to increased prevalence of tyrosine decarboxylase (Tdc)-expressing bacteria and resulting tyramine production. Tyramine acts on the tyramine receptor, TyrR1, to promote cytosolic Ca2+ signaling and activation of the CRTC-CREB complex to transcriptionally suppress dietary lipid digestion and lipogenesis in enterocytes, while promoting mitochondrial biogenesis. Furthermore, the tyramine-induced cytosolic Ca2+ signaling is sufficient to suppress HFD-induced obesity and insulin resistance in Drosophila. In mice, tyramine intake also improves glucose tolerance and insulin sensitivity under HFD. These results indicate that dysbiosis-induced tyramine suppresses insulin resistance in both flies and mice under HFD, suggesting a potential therapeutic strategy for related metabolic disorders, such as diabetes.


Subject(s)
Calcium Signaling , Diet, High-Fat , Gastrointestinal Microbiome , Insulin Resistance , Tyramine , Animals , Tyramine/metabolism , Tyramine/pharmacology , Gastrointestinal Microbiome/drug effects , Diet, High-Fat/adverse effects , Mice , Calcium Signaling/drug effects , Obesity/metabolism , Obesity/microbiology , Obesity/etiology , Male , Drosophila/metabolism , Dysbiosis/metabolism , Dysbiosis/microbiology , Mice, Inbred C57BL , Drosophila melanogaster/microbiology , Drosophila melanogaster/metabolism , Enterocytes/metabolism , Enterocytes/drug effects
2.
Eur J Pharm Biopharm ; 202: 114408, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39004319

ABSTRACT

The therapeutic effects of orally administered nanocarriers depend on their ability to effectively permeate the intestinal mucosa, which is one of the major challenges in oral drug delivery. Microfold cells are specialized enterocytes in the intestinal epithelium known for their high transcytosis abilities. This study aimed to compare and evaluate two targeting approaches using surface modifications of polymer-based nanocarriers, whereas one generally addresses enterocytes, and one is directed explicitly to microfold cells via targeting the sialyl LewisA motif on their surface. We characterized the resulting carriers in terms of size and charge, supplemented by scanning electron microscopy to confirm their structural properties. For predictive biological testing and to assess the intended targeting effect, we implemented two human intestinal in vitro models containing microfold-like cells. Both models were thoroughly characterized prior to permeation studies with the different nanocarriers. Our results demonstrated improved transport for both targeted formulations compared to undecorated carriers in the in vitro models. Notably, there was an enhanced uptake in the presence of microfold-like cells, particularly for the nanocarriers directed by the anti-sialyl LewisA antibody. These findings highlight the potential of microfold cell targeting to improve oral administration of drugs and emphasize the importance of using suitable and well-characterized in vitro models for testing novel drug delivery strategies.


Subject(s)
Drug Carriers , Drug Delivery Systems , Intestinal Mucosa , M Cells , Nanoparticles , Humans , Administration, Oral , Caco-2 Cells , Drug Carriers/chemistry , Drug Delivery Systems/methods , Enterocytes/metabolism , Enterocytes/drug effects , Intestinal Absorption/drug effects , Intestinal Mucosa/metabolism , M Cells/metabolism , Nanoparticles/chemistry , Permeability , Polymers/chemistry
3.
Biomolecules ; 14(6)2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38927069

ABSTRACT

The management of gastrointestinal disease in animals represents a significant challenge in veterinary and zootechnic practice. Traditionally, acute symptoms have been treated with antibiotics and high doses of zinc oxide (ZnO). However, concerns have been raised regarding the potential for microbial resistance and ecological detriment due to the excessive application of this compound. These concerns highlight the urgency of minimizing the use of ZnO and exploring sustainable nutritional solutions. Hydrolysable tannins (HTs), which are known for their role in traditional medicine for acute gastrointestinal issues, have emerged as a promising alternative. This study examined the combined effect of food-grade HTs and subtherapeutic ZnO concentration on relevant biological functions of Caco-2 cells, a widely used model of the intestinal epithelial barrier. We found that, when used together, ZnO and HTs (ZnO/HTs) enhanced tissue repair and improved epithelial barrier function, normalizing the expression and functional organization of tight junction proteins. Finally, the ZnO/HTs combination strengthened enterocytes' defense against oxidative stress induced by inflammation stimuli. In conclusion, combining ZnO and HTs may offer a suitable and practical approach for decreasing ZnO levels in veterinary nutritional applications.


Subject(s)
Enterocytes , Hydrolyzable Tannins , Zinc Oxide , Zinc Oxide/pharmacology , Zinc Oxide/chemistry , Caco-2 Cells , Enterocytes/drug effects , Enterocytes/metabolism , Humans , Hydrolyzable Tannins/pharmacology , Hydrolyzable Tannins/chemistry , Oxidative Stress/drug effects , Tight Junction Proteins/metabolism
4.
Cell Death Dis ; 15(4): 301, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38684650

ABSTRACT

Understanding the mechanisms involved in colonic epithelial differentiation is key to unraveling the alterations causing inflammatory conditions and cancer. Organoid cultures provide an unique tool to address these questions but studies are scarce. We report a differentiation system toward enterocytes and goblet cells, the two major colonic epithelial cell lineages, using colon organoids generated from healthy tissue of colorectal cancer patients. Culture of these organoids in medium lacking stemness agents resulted in a modest ultrastructural differentiation phenotype with low-level expression of enterocyte (KLF4, KRT20, CA1, FABP2) and goblet cell (TFF2, TFF3, AGR2) lineage markers. BMP pathway activation through depletion of Noggin and addition of BMP4 resulted in enterocyte-biased differentiation. Contrarily, blockade of the Notch pathway using the γ-secretase inhibitor dibenzazepine (DBZ) favored goblet cell differentiation. Combination treatment with BMP4 and DBZ caused a balanced strong induction of both lineages. In contrast, colon tumor organoids responded poorly to BMP4 showing only weak signals of cell differentiation, and were unresponsive to DBZ. We also investigated the effects of 1α,25-dihydroxyvitamin D3 (calcitriol) on differentiation. Calcitriol attenuated the effects of BMP4 and DBZ on colon normal organoids, with reduced expression of differentiation genes and phenotype. Consistently, in normal organoids, calcitriol inhibited early signaling by BMP4 as assessed by reduction of the level of phospho-SMAD1/5/8. Our results show that BMP and Notch signaling play key roles in human colon stem cell differentiation to the enterocytic and goblet cell lineages and that calcitriol modulates these processes favoring stemness features.


Subject(s)
Bone Morphogenetic Protein 4 , Calcitriol , Carrier Proteins , Cell Differentiation , Colon , Dibenzazepines , Goblet Cells , Kruppel-Like Factor 4 , Organoids , Receptors, Notch , Signal Transduction , Humans , Organoids/drug effects , Organoids/metabolism , Cell Differentiation/drug effects , Bone Morphogenetic Protein 4/metabolism , Colon/drug effects , Colon/metabolism , Colon/cytology , Colon/pathology , Receptors, Notch/metabolism , Signal Transduction/drug effects , Calcitriol/pharmacology , Goblet Cells/drug effects , Goblet Cells/metabolism , Dibenzazepines/pharmacology , Cell Lineage/drug effects , Enterocytes/metabolism , Enterocytes/drug effects , Enterocytes/cytology , Vitamin D/pharmacology
5.
Res Vet Sci ; 154: 132-137, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36584521

ABSTRACT

Several factors such as pathogen bacteria, or oral chemotherapy disturb the intestinal integrity, leading to several undesirable effects. Inactivated probiotics may be beneficial in safely redress the physiological functions of the intestinal epithelium. Our aim is to determine the effect of tyndallized Lactobacillus on LPS- and 5-fluorouracil-treated porcine jejunal cells. IPEC-J2 cells derived from porcine jejunal epithelium were used as the in vitro model. The enterocyte cell cultures were treated with 109Lactobacillus reuteri cells/ml or 10 µg/ml lipopolysaccharides (LPS) or 100 µM 5-fluorouracil separately and simultaneously. We determined the alterations in mRNA levels of inflammatory mediators IL6, CXCL8/IL8, TNF. Furthermore, the protein level of IL-6 and IL-8 were measured. The fluorouracil treatment upregulated the IL6 gene expression, the endotoxin treatment upregulated the IL8 and TNF level. The heat-inactivated Lactobacillus increased the IL-8 level both at the gene expression and protein level. The co-administration of the non-viable probiotic with the 5-fluorouracil and the LPS resulted in decrease of IL6, IL8, and TNF level. The immune-modulator effect of tyndallized probiotic product is demonstrated in porcine jejunal cells. The inactivated Lactobacillus was able to prevent the accumulation of the selected inflammatory mediators in LPS- or 5-fluorouracil-exposed enterocytes.


Subject(s)
Enterocytes , Probiotics , Animals , Endotoxins , Enterocytes/drug effects , Hot Temperature , Interleukin-6/genetics , Interleukin-8/genetics , Interleukin-8/metabolism , Intestinal Mucosa/metabolism , Lactobacillus/physiology , Lipopolysaccharides , Probiotics/pharmacology , Swine , Fluorouracil
6.
Carbohydr Polym ; 291: 119655, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-35698356

ABSTRACT

Roots of Aconitum carmichaelii are used in Asian countries due to its content of bioactive alkaloids. In the production of root preparations, tons of leaves are usually discarded, leading to a huge waste of herbal material. The aim of this study is to investigate the polysaccharides in these unutilized leaves. A neutral polysaccharide (AL-N) appeared to be a mixture of heteromannans, and two purified acidic polysaccharides (AL-I-I and AL-I-II) were shown to be pectins containing a homogalacturonan backbone substituted with terminal ß-Xylp-units. AL-I-I consisted of a type-I rhamnogalacturonan core, with arabinan and type-II arabinogalactan domains while AL-I-II was less branched. AL-N and AL-I-I were able to modulate the complement system, while AL-I-II was inactive. Interestingly, AL-N, AL-I-I and AL-I-II were shown to exert anti-inflammatory effects on porcine enterocyte IPEC-J2 cells. AL-I-I and AL-I-II were able to down-regulate the expression of toll-like receptor 4 (TLR4) and nucleotide-binding oligomerization domain 1 (NOD1).


Subject(s)
Aconitum/chemistry , Alkaloids , Polysaccharides , Alkaloids/analysis , Alkaloids/chemistry , Animals , Anti-Inflammatory Agents/analysis , Anti-Inflammatory Agents/pharmacology , Cell Line , Enterocytes/drug effects , Plant Leaves/chemistry , Polysaccharides/analysis , Polysaccharides/chemistry , Polysaccharides/pharmacology , Swine
7.
J Nutr Biochem ; 108: 109087, 2022 10.
Article in English | MEDLINE | ID: mdl-35691593

ABSTRACT

Although the role of mechanistic target of rapamycin complex 1 (mTORC1) in lipid metabolism has been the subject of previous research, its function in chylomicron production is not known. In this study, we created three stable human colorectal adenocarcinoma Caco-2 cell lines exhibiting normal, low, or high mTORC1 kinase activity, and used these cells to investigate the consequences of manipulating mTORC1 activity on enterocyte differentiation and chylomicron-like particle production. Constitutively active mTORC1 induced Caco-2 cell proliferation and differentiation (as judged by alkaline phosphatase activity) but weakened transepithelial electrical resistance (TEER). Repressed mTORC1 activity due to the knockdown of RPTOR significantly decreased the expression of lipogenic genes FASN, DGAT1, and DGAT2, lipoprotein assembly genes APOB and MTTP, reduced protein expression of APOB, MTTP, and FASN, downregulated the gene expression of very long-chain fatty acyl-CoA ligase (FATP2), acyl-CoA binding protein (DBI), and prechylomicron transport vesicle-associated proteins VAMP7 (vesicle-associated membrane protein 7) and SAR1B (secretion associated Ras related GTPase 1B) resulting in the repression of apoB-containing triacylglycerol-rich lipoprotein secretion. Exposure of Caco-2 cells harboring a constitutively active mTORC1 to short-chain fatty acid derivatives, R-α-lipoic acid and 4-phenylbutyric acid, downregulated chylomicron-like particle secretion by interfering with the lipidation and assembly of the particles, and concomitantly repressed mTORC1 activity with no change to Raptor abundance or PRAS40 (Thr246) phosphorylation. R-α-lipoic acid and 4-phenylbutyric acid may be useful to mitigate intestinal lipoprotein overproduction and associated postprandial inflammation.


Subject(s)
Chylomicrons , Enterocytes , Monomeric GTP-Binding Proteins , Phenylbutyrates , Regulatory-Associated Protein of mTOR , Thioctic Acid , Apolipoproteins B/metabolism , Caco-2 Cells , Chylomicrons/metabolism , Coenzyme A Ligases/metabolism , Enterocytes/drug effects , Enterocytes/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Phenylbutyrates/metabolism , Phenylbutyrates/pharmacology , Regulatory-Associated Protein of mTOR/genetics , Regulatory-Associated Protein of mTOR/metabolism , Thioctic Acid/metabolism , Thioctic Acid/pharmacology
8.
Molecules ; 27(3)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35164139

ABSTRACT

Butyrate is a major gut microbiome metabolite that regulates several defense mechanisms against infectious diseases. Alterations in the gut microbiome, leading to reduced butyrate production, have been reported in patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. A new butyrate releaser, useful for all the known applications of butyrate, presenting physiochemical characteristics suitable for easy oral administration, (N-(1-carbamoyl-2-phenyl-ethyl) butyramide (FBA), has been recently developed. We investigated the protective action of FBA against SARS-CoV-2 infection in the human small intestine and enterocytes. Relevant aspects of SARS-CoV-2 infection were assessed: infectivity, host functional receptor angiotensin-converting enzyme-2 (ACE2), transmembrane protease serine 2 (TMPRSS2), neuropilin-1 (NRP1), pro-inflammatory cytokines expression, genes involved in the antiviral response and the activation of Nf-kB nuclear factor (erythroid-derived 2-like) 2 (Nfr2) pathways. We found that FBA positively modulates the crucial aspects of the infection in small intestinal biopsies and human enterocytes, reducing the expression of ACE2, TMPRSS2 and NRP1, pro-inflammatory cytokines interleukin (IL)-15, monocyte chemoattractant protein-1 (MCP-1) and TNF-α, and regulating several genes involved in antiviral pathways. FBA was also able to reduce the number of SARS-CoV-2-infected cells, and ACE2, TMPRSS2 and NRP1 expression. Lastly, through the inhibition of Nf-kB and the up-regulation of Nfr2, it was also able to reduce the expression of pro-inflammatory cytokines IL-15, MCP-1 and TNF-α in human enterocytes. The new butyrate releaser, FBA, exerts a preventive action against SARS-CoV-2 infection. It could be considered as an innovative strategy to limit COVID-19.


Subject(s)
Butyrates/pharmacology , COVID-19 Drug Treatment , SARS-CoV-2/metabolism , Antiviral Agents/pharmacology , Butyrates/metabolism , COVID-19/metabolism , Caco-2 Cells , Enterocytes/drug effects , Enterocytes/metabolism , Gene Expression/genetics , Gene Expression Regulation/genetics , Humans , Intestines/drug effects , Intestines/metabolism , Male , SARS-CoV-2/drug effects , SARS-CoV-2/pathogenicity
9.
Cell Rep ; 37(13): 110150, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34965418

ABSTRACT

Enteric pathogens overcome barrier immunity within the intestinal environment that includes the endogenous flora. The microbiota produces diverse ligands, and the full spectrum of microbial products that are sensed by the epithelium and prime protective immunity is unknown. Using Drosophila, we find that the gut presents a high barrier to infection, which is partially due to signals from the microbiota, as loss of the microbiota enhances oral viral infection. We report cyclic dinucleotide (CDN) feeding is sufficient to protect microbiota-deficient flies from enhanced oral infection, suggesting that bacterial-derived CDNs induce immunity. Mechanistically, we find CDN protection is dSTING- and dTBK1-dependent, leading to NF-kB-dependent gene expression. Furthermore, we identify the apical nucleoside transporter, CNT2, as required for oral CDN protection. Altogether, our studies define a role for bacterial products in priming immune defenses in the gut.


Subject(s)
Alphavirus Infections/immunology , Antiviral Agents/pharmacology , Drosophila melanogaster/immunology , Enterocytes/immunology , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Nucleotides, Cyclic/administration & dosage , Alphavirus Infections/drug therapy , Alphavirus Infections/virology , Animals , Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Enterocytes/drug effects , Enterocytes/virology , Female , Immunity, Innate , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sindbis Virus/immunology
10.
Nutrients ; 13(9)2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34579029

ABSTRACT

Ferulic acid (FA) is a polyphenol pertaining to the class of hydroxycinnamic acids present in numerous foods of a plant origin. Its dietary consumption leads to the formation of several phase I and II metabolites in vivo, which represent the largest amount of ferulates in the circulation and in the intestine in comparison with FA itself. In this work, we evaluated their efficacy against the proinflammatory effects induced by lipopolysaccharide (LPS) in intestinal Caco-2 cell monolayers, as well as the mechanisms underlying their protective action. LPS-induced overexpression of proinflammatory enzymes such as inducible nitric oxide synthase (iNOS) and the consequent hyperproduction of nitric oxide (NO) and cyclic guanosine monophosphate (cGMP) were limited by physiological relevant concentrations (1 µM) of FA, its derivatives isoferulic acid (IFA) and dihydroferulic acid (DHFA), and their glucuronidated and sulfated metabolites, which acted upstream by limiting the activation of MAPK p38 and ERK and of Akt kinase, thus decreasing the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-ĸB) translocation into the nucleus. Furthermore, the compounds were found to promote the expression of Nrf2, which may have contributed to the downregulation of NF-ĸB activity. The overall data show that phase I/II metabolites retain the efficacy of their dietary free form in contrasting inflammatory response.


Subject(s)
Coumaric Acids/pharmacology , Enterocytes/drug effects , Inflammation/chemically induced , Lipopolysaccharides/toxicity , Caco-2 Cells , Cell Survival/drug effects , Coumaric Acids/metabolism , Cyclic GMP/genetics , Cyclic GMP/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Inflammation/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Proto-Oncogene Proteins c-akt , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Int J Biol Macromol ; 189: 410-419, 2021 Oct 31.
Article in English | MEDLINE | ID: mdl-34437917

ABSTRACT

We have previously demonstrated the ability of the human vaginal strain Lactobacillus crispatus 2029 (LC2029) for strong adhesion to cervicovaginal epithelial cells, expression of the surface layer protein 2 (Slp2), and antagonistic activity against urogenital pathogens. Slp2 forms regular two-dimensional structure around the LC2029 cells,which is secreted into the medium and inhibits intestinal pathogen-induced activation of caspase-9 and caspase-3 in the human intestinal Caco-2 cells. Here, we elucidated the effects of soluble Slp2 on adhesion of proteobacteria pathogens inducing necrotizing enterocolitis (NEC), such as Escherichia coli ATCC E 2348/69, E. coli ATCC 31705, Salmonella Enteritidis ATCC 13076, Campylobacter jejuni ATCC 29428, and Pseudomonas aeruginosa ATCC 27853 to Caco-2 cells, as well as on growth promotion, differentiation, vascular endothelial growth factor (VEGF) production, and intestinal barrier function of Caco-2 cell monolayers. Slp2 acts as anti-adhesion agent for NEC-inducing proteobacteria, promotes growth of immature Caco-2 cells and their differentiation, and enhances expression and functional activity of sucrase, lactase, and alkaline phosphatase. Slp2 stimulates VEGF production, decreases paracellular permeability, and increases transepithelial electrical resistance, strengthening barrier function of Caco-2 cell monolayers. These data support the important role of Slp2 in the early postnatal development of the human small intestine enterocytes.


Subject(s)
Cell Differentiation , Enterocytes/metabolism , Lactobacillus crispatus/chemistry , Membrane Glycoproteins/pharmacology , Vagina/microbiology , Vascular Endothelial Growth Factor A/biosynthesis , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Bacterial Adhesion/drug effects , Caco-2 Cells , Cell Differentiation/drug effects , Cell Membrane Permeability/drug effects , Cell Proliferation/drug effects , Electric Impedance , Enterocytes/drug effects , Female , Gene Expression Regulation, Enzymologic/drug effects , Humans , Lactase/genetics , Lactase/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sucrase/genetics , Sucrase/metabolism
12.
Oxid Med Cell Longev ; 2021: 8936907, 2021.
Article in English | MEDLINE | ID: mdl-34373770

ABSTRACT

Probiotics are widely used for protection against stress-induced intestinal dysfunction. Oxidative stress plays a critical role in gastrointestinal disorders. It is established that probiotics alleviate oxidative stress; however, the mechanism of action has not been elucidated. We developed an in vitro intestinal porcine epithelial cells (IPEC-J2) model of oxidative stress to explore the antioxidant effect and potential mode of action of Lactobacillus plantarum ZLP001. The IPEC-J2 cells were preincubated with and without L. plantarum ZLP001 for 3 h and then exposed to hydrogen peroxide (H2O2) for 4 h. Pretreatment with L. plantarum ZLP001 protected IPEC-J2 cells against H2O2-induced oxidative damage as indicated by cell viability assays and significantly alleviated apoptosis elicited by H2O2. L. plantarum ZLP001 pretreatment decreased reactive oxygen species production and the cellular malondialdehyde concentration and increased the mitochondrial membrane potential compared with H2O2 treatment alone, suggesting that L. plantarum ZLP001 promotes the maintenance of redox homeostasis in the cells. Furthermore, L. plantarum ZLP001 regulated the expression and generation of some antioxidant enzymes, thereby activating the antioxidant defense system. Treatment with L. plantarum ZLP001 led to nuclear erythroid 2-related factor 2 (Nrf2) enrichment in the nucleus compared with H2O2 treatment alone. Knockdown of Nrf2 significantly weakened the alleviating effect of L. plantarum ZLP001 on antioxidant stress in IPEC-J2 cells, suggesting that Nrf2 is involved in the antioxidative effect of L. plantarum ZLP001. Collectively, these results indicate that L. plantarum ZLP001 is a promising probiotic bacterium that can potentially alleviate oxidative stress.


Subject(s)
Enterocytes/metabolism , Gastrointestinal Microbiome , Lactobacillus plantarum/pathogenicity , Oxidative Stress , Animals , Cells, Cultured , Enterocytes/drug effects , Enterocytes/microbiology , Hydrogen Peroxide/toxicity , Ileum/cytology , Ileum/metabolism , Ileum/microbiology , Swine
13.
Eur J Pharmacol ; 909: 174408, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34364877

ABSTRACT

The cellular and tissue damage induced by oxidative stress (OS) contribute to a variety of human diseases, which include gastrointestinal diseases. Salvianolic acid B (Sal B), which is a natural polyphenolic acid in Salvia miltiorrhiza, exhibits prominent antioxidant properties. However, its precise function and molecular mechanisms in protecting normal intestine epithelium from OS-induced damage are still poorly defined. In this study, we tried to clarify this relationship. Here, we found Sal B addiction in the rat intestinal epithelial cell, IEC-6, prevented H2O2-induced cell viability decrease and apoptosis induction, ameliorated H2O2-induced intestinal epithelial barrier dysfunction and mitochondrial dysfunction, and suppressed H2O2-induced production of ROS to varying degrees, ranging from 10% to 30%. Moreover, by employing an ischemia reperfusion model of rats, we also discovered that Sal B treatment reversed ischemia and a reperfusion-caused decrease in villus height and crypt depth, decreased proliferation of enterocytes, and increased the apoptotic index in the jejunum and ileum. Mechanistically, Sal B treatment up-regulated the phosphorylated level of Akt and GSK3ß in enterocytes in vitro and in vivo, and PI3K inhibitor LY294002 treatment abrogated the protective effects of Sal B. Meanwhile, the inactivation of GSK3ß reversed the oxidative stress-induced apoptosis and mitochondrial dysfunction in IEC-6 cells. Together, our results demonstrated that the damage of intestinal epithelial cells in in vitro and in vivo models were both attenuated by Sal B treatment, and such antioxidant activity might very possibly be attributed to the activation of Akt/GSK3ß signaling.


Subject(s)
Antioxidants/pharmacology , Benzofurans/pharmacology , Intestinal Diseases/drug therapy , Animals , Antioxidants/therapeutic use , Apoptosis/drug effects , Benzofurans/therapeutic use , Cell Line , Disease Models, Animal , Enterocytes/drug effects , Enterocytes/pathology , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Ileum/cytology , Ileum/drug effects , Ileum/pathology , Intestinal Diseases/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Jejunum/cytology , Jejunum/drug effects , Jejunum/pathology , Male , Mitochondria/drug effects , Mitochondria/pathology , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Salvia miltiorrhiza/chemistry , Signal Transduction/drug effects
14.
Cell Death Dis ; 12(9): 815, 2021 08 27.
Article in English | MEDLINE | ID: mdl-34453041

ABSTRACT

Crohn's disease (CD) is an intestinal immune-dysfunctional disease. Extracellular vesicles (EVs) are membrane-enclosed particles full of functional molecules, e.g., nuclear acids. Recently, EVs have been shown to participate in the development of CD by realizing intercellular communication among intestinal cells. However, the role of EVs carrying double-strand DNA (dsDNA) shed from sites of intestinal inflammation in CD has not been investigated. Here we isolated EVs from the plasma or colon lavage of murine colitis and CD patients. The level of exosomal dsDNA, including mtDNA and nDNA, significantly increased in murine colitis and active human CD, and was positively correlated with the disease activity. Moreover, the activation of the STING pathway was verified in CD. EVs from the plasma of active human CD triggered STING activation in macrophages in vitro. EVs from LPS-damaged colon epithelial cells were also shown to raise inflammation in macrophages via activating the STING pathway, but the effect disappeared after the removal of exosomal dsDNA. These findings were further confirmed in STING-deficient mice and macrophages. STING deficiency significantly ameliorated colitis. Besides, potential therapeutic effects of GW4869, an inhibitor of EVs release were assessed. The application of GW4869 successfully ameliorated murine colitis by inhibiting STING activation. In conclusion, exosomal dsDNA was found to promote intestinal inflammation via activating the STING pathway in macrophages and act as a potential mechanistic biomarker and therapeutic target of CD.


Subject(s)
Crohn Disease/pathology , DNA/metabolism , Extracellular Vesicles/metabolism , Membrane Proteins/metabolism , Signal Transduction , Aniline Compounds/pharmacology , Animals , Benzylidene Compounds/pharmacology , Colitis/pathology , Disease Models, Animal , Endocytosis/drug effects , Enterocytes/drug effects , Enterocytes/pathology , Enterocytes/ultrastructure , Extracellular Vesicles/drug effects , Extracellular Vesicles/ultrastructure , Humans , Inflammation/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/ultrastructure , Membrane Proteins/deficiency , Mice, Knockout , Models, Biological , Phosphorylation/drug effects , Signal Transduction/drug effects
15.
Am J Physiol Cell Physiol ; 321(3): C471-C488, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34288721

ABSTRACT

Intestinal NaCl, HCO3-, and fluid absorption are strongly dependent on apical Na+/H+ exchange. The intestine expresses three presumably apical sodium-hydrogen exchanger (NHE) isoforms: NHE2, NHE3, and NHE8. We addressed the role of NHE8 [solute carrier 9A8 (SLC9A8)] and its interplay with NHE2 (SLC9A2) in luminal proton extrusion during acute and chronic enterocyte acidosis and studied the differential effects of NHE8 and NHE2 on enterocyte proliferation. In contrast to NHE3, which was upregulated in differentiated versus undifferentiated colonoids, the expression of NHE2 and NHE8 remained constant during differentiation of colonoids and Caco2Bbe cells. Heterogeneously expressed Flag-tagged rat (r)Nhe8 and human (h)NHE8 translocated to the apical membrane of Caco2Bbe cells. rNhe8 and hNHE8, when expressed in NHE-deficient PS120 fibroblasts showed higher sensitivity to HOE642 compared to NHE2. Lentiviral shRNA knockdown of endogenous NHE2 in Caco2Bbe cells (C2Bbe/shNHE2) resulted in a decreased steady-state intracellular pH (pHi), an increased NHE8 mRNA expression, and augmented NHE8-mediated apical NHE activity. Lentiviral shRNA knockdown of endogenous NHE8 in Caco2Bbe cells (C2Bbe/shNHE8) resulted in a decreased steady-state pHi as well, accompanied by decreased NHE2 mRNA expression and activity, which together contributed to reduced apical NHE activity in the NHE8-knockdown cells. Chronic acidosis increased NHE8 but not NHE2 mRNA expression. Alterations in NHE2 and NHE8 expression/activity affected proliferation, with C2Bbe/shNHE2 cells having lower and C2Bbe/shNHE8 having higher proliferative capacity, accompanied by amplified ERK1/2 signaling pathway and increased EGFR expression in the latter cell line. Thus, both Na+/H+ exchangers have distinct functions during cellular homeostasis by triggering different signaling pathways to regulate cellular proliferation and pHi control.


Subject(s)
Colon/metabolism , Enterocytes/metabolism , Sodium-Hydrogen Exchangers/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Colon/cytology , Colon/drug effects , Enterocytes/cytology , Enterocytes/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Guanidines/pharmacology , HT29 Cells , Homeostasis/genetics , Humans , Hydrogen-Ion Concentration , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Organoids/cytology , Organoids/drug effects , Organoids/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Signal Transduction , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/metabolism , Sulfones/pharmacology
16.
JCI Insight ; 6(16)2021 08 23.
Article in English | MEDLINE | ID: mdl-34197342

ABSTRACT

Functional loss of myosin Vb (MYO5B) induces a variety of deficits in intestinal epithelial cell function and causes a congenital diarrheal disorder, microvillus inclusion disease (MVID). The impact of MYO5B loss on differentiated cell lineage choice has not been investigated. We quantified the populations of differentiated epithelial cells in tamoxifen-induced, epithelial cell-specific MYO5B-knockout (VilCreERT2 Myo5bfl/fl) mice utilizing digital image analysis. Consistent with our RNA-sequencing data, MYO5B loss induced a reduction in tuft cells in vivo and in organoid cultures. Paneth cells were significantly increased by MYO5B deficiency along with expansion of the progenitor cell zone. We further investigated the effect of lysophosphatidic acid (LPA) signaling on epithelial cell differentiation. Intraperitoneal LPA significantly increased tuft cell populations in both control and MYO5B-knockout mice. Transcripts for Wnt ligands were significantly downregulated by MYO5B loss in intestinal epithelial cells, whereas Notch signaling molecules were unchanged. Additionally, treatment with the Notch inhibitor dibenzazepine (DBZ) restored the populations of secretory cells, suggesting that the Notch pathway is maintained in MYO5B-deficient intestine. MYO5B loss likely impairs progenitor cell differentiation in the small intestine in vivo and in vitro, partially mediated by Wnt/Notch imbalance. Notch inhibition and/or LPA treatment may represent an effective therapeutic approach for treatment of MVID.


Subject(s)
Malabsorption Syndromes/genetics , Microvilli/pathology , Mucolipidoses/genetics , Myosin Type V/deficiency , Receptors, Notch/metabolism , Wnt Signaling Pathway/genetics , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Dibenzazepines/pharmacology , Disease Models, Animal , Enterocytes/drug effects , Enterocytes/metabolism , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Jejunum/cytology , Jejunum/drug effects , Jejunum/pathology , Lysophospholipids/pharmacology , Lysophospholipids/therapeutic use , Malabsorption Syndromes/drug therapy , Malabsorption Syndromes/pathology , Mice , Mice, Knockout , Microvilli/genetics , Mucolipidoses/drug therapy , Mucolipidoses/pathology , Myosin Type V/genetics , Organoids , Primary Cell Culture , Receptors, Notch/antagonists & inhibitors , Stem Cells/physiology , Wnt Signaling Pathway/drug effects
17.
Cell Mol Gastroenterol Hepatol ; 12(3): 943-981, 2021.
Article in English | MEDLINE | ID: mdl-34102314

ABSTRACT

BACKGROUND & AIMS: The use of antibiotics (ABs) is a common practice during the first months of life. ABs can perturb the intestinal microbiota, indirectly influencing the intestinal epithelial cells (IECs), but can also directly affect IECs independent of the microbiota. Previous studies have focused mostly on the impact of AB treatment during adulthood. However, the difference between the adult and neonatal intestine warrants careful investigation of AB effects in early life. METHODS: Neonatal mice were treated with a combination of amoxicillin, vancomycin, and metronidazole from postnatal day 10 to 20. Intestinal permeability and whole-intestine gene and protein expression were analyzed. IECs were sorted by a fluorescence-activated cell sorter and their genome-wide gene expression was analyzed. Mouse fetal intestinal organoids were treated with the same AB combination and their gene and protein expression and metabolic capacity were determined. RESULTS: We found that in vivo treatment of neonatal mice led to decreased intestinal permeability and a reduced number of specialized vacuolated cells, characteristic of the neonatal period and necessary for absorption of milk macromolecules. In addition, the expression of genes typically present in the neonatal intestinal epithelium was lower, whereas the adult gene expression signature was higher. Moreover, we found altered epithelial defense and transepithelial-sensing capacity. In vitro treatment of intestinal fetal organoids with AB showed that part of the consequences observed in vivo is a result of the direct action of the ABs on IECs. Lastly, ABs reduced the metabolic capacity of intestinal fetal organoids. CONCLUSIONS: Our results show that early life AB treatment induces direct and indirect effects on IECs, influencing their maturation and functioning.


Subject(s)
Amoxicillin/administration & dosage , Anti-Bacterial Agents/administration & dosage , Gene Regulatory Networks/drug effects , Intestines/metabolism , Metronidazole/administration & dosage , Vancomycin/administration & dosage , Amoxicillin/adverse effects , Animals , Animals, Newborn , Anti-Bacterial Agents/adverse effects , Disease Models, Animal , Enterocytes/cytology , Enterocytes/drug effects , Enterocytes/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Intestines/cytology , Intestines/drug effects , Metronidazole/adverse effects , Mice , Oligonucleotide Array Sequence Analysis , Permeability/drug effects , Postnatal Care , Vacuoles/drug effects , Vacuoles/metabolism , Vancomycin/adverse effects
18.
Mol Biol Rep ; 48(5): 4667-4675, 2021 May.
Article in English | MEDLINE | ID: mdl-34023987

ABSTRACT

The transmembrane protease serine 2 (TMPRSS2) is a membrane anchored protease that primarily expressed by epithelial cells of respiratory and gastrointestinal systems and has been linked to multiple pathological processes in humans including tumor growth, metastasis and viral infections. Recent studies have shown that TMPRSS2 expressed on cell surface of host cells could play a crucial role in activation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein which facilitates the rapid early entry of the virus into host cells. In addition, direct suppression of TMPRSS2 using small drug inhibitors has been demonstrated to be effective in decreasing SARS-CoV-2 infection in vitro, which presents TMPRSS2 protease as a potential therapeutic strategy for SARS-CoV-2 infection. Recently, SARS-CoV-2 has been shown to be capable of infecting gastrointestinal enterocytes and to provoke gastrointestinal disorders in patients with COVID-19 disease, which is considered as a new transmission route and target organ of SARS-CoV-2. In this review, we highlight the biochemical properties of TMPRSS2 protease and discuss the potential targeting of TMPRSS2 by inhibitors to prevent the SARS-CoV-2 spreading through gastro-intestinal tract system as well as the hurdles that need to be overcome.


Subject(s)
COVID-19/metabolism , Enterocytes/drug effects , SARS-CoV-2/physiology , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology , Antiviral Agents/pharmacology , Drug Evaluation, Preclinical , Enterocytes/metabolism , Enterocytes/virology , Humans , SARS-CoV-2/drug effects , Small Molecule Libraries/pharmacology , Spike Glycoprotein, Coronavirus/metabolism , Virus Internalization/drug effects , COVID-19 Drug Treatment
19.
J Radiat Res ; 62(4): 574-581, 2021 Jul 10.
Article in English | MEDLINE | ID: mdl-33912959

ABSTRACT

Intrinsic autophagy is important for the maintenance of intestinal homeostasis and intestinal regeneration. Ionizing radiation suppresses intrinsic autophagy and reduces damage-induced regeneration in the intestine, resulting in intestinal injury. Resveratrol, a sirtuin 1 (SIRT1) agonist, promotes autophagy and exerts radioprotective effect. In this study, the protective effect of resveratrol against radiation-induced intestinal injury and its potential mechanism were investigated. Intestinal epithelial cells (IEC-6) were exposed to 10 Gy ionizing radiation and resveratrol (0.1-40.0 µM). Cell viability was investigated using Cell Counting Kit 8 (CCK8), apoptosis was observed by Annexin V-fluorescein isothiocyanate/propidium iodide (PI) staining and flow cytometry, and the expression of apoptotic and autophagic proteins was determined by western blotting. Resveratrol exerted a high toxicity against IEC-6 cells, but at low concentrations, it inhibited ionizing radiation-induced apoptosis. Resveratrol increased SIRT1 expression after irradiation and inhibited ionizing radiation-induced p53 acetylation and pro-apoptotic protein, Bax, expression. Furthermore, resveratrol promoted autophagy via the phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway, thereby protecting IEC-6 cells against radiation-induced damage. These results suggest that resveratrol reduces radiation-induced IEC-6 cell damage by inhibiting apoptosis and promoting autophagy via the activation of SIRT1, and that the PI3K/AKT/mTOR signaling pathway is involved in the induction of autophagy.


Subject(s)
Apoptosis , Autophagy , Cytoprotection , Enterocytes/pathology , Radiation, Ionizing , Resveratrol/pharmacology , Sirtuin 1/metabolism , Acetylation/drug effects , Acetylation/radiation effects , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Autophagy/drug effects , Autophagy/radiation effects , Beclin-1/metabolism , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , Cytoprotection/drug effects , Cytoprotection/radiation effects , Enterocytes/drug effects , Enterocytes/radiation effects , Mice , Microtubule-Associated Proteins/metabolism , Models, Biological , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism
20.
Int J Mol Sci ; 22(8)2021 Apr 17.
Article in English | MEDLINE | ID: mdl-33920650

ABSTRACT

Electrolytes (NaCl) and fluid malabsorption cause diarrhea in inflammatory bowel disease (IBD). Coupled NaCl absorption, mediated by Na+/H+ and Cl-/HCO3- exchanges on the intestinal villus cells brush border membrane (BBM), is inhibited in IBD. Arachidonic acid metabolites (AAMs) formed via cyclooxygenase (COX) or lipoxygenase (LOX) pathways are elevated in IBD. However, their effects on NaCl absorption are not known. We treated SAMP1/YitFc (SAMP1) mice, a model of spontaneous ileitis resembling human IBD, with Arachidonyl Trifluoro Methylketone (ATMK, AAM inhibitor), or with piroxicam or MK-886, to inhibit COX or LOX pathways, respectively. Cl-/HCO3- exchange, measured as DIDS-sensitive 36Cl uptake, was significantly inhibited in villus cells and BBM vesicles of SAMP1 mice compared to AKR/J controls, an effect reversed by ATMK. Piroxicam, but not MK-886, also reversed the inhibition. Kinetic studies showed that inhibition was secondary to altered Km with no effects on Vmax. Whole cell or BBM protein levels of Down-Regulated in Adenoma (SLC26A3) and putative anion transporter-1 (SLC26A6), the two key BBM Cl-/HCO3- exchangers, were unaltered. Thus, inhibition of villus cell Cl-/HCO3- exchange by COX pathway AAMs, such as prostaglandins, via reducing the affinity of the exchanger for Cl-, and thereby causing NaCl malabsorption, could significantly contribute to IBD-associated diarrhea.


Subject(s)
Arachidonic Acids/metabolism , Chloride-Bicarbonate Antiporters/metabolism , Enterocytes/metabolism , Ileitis/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Arachidonic Acids/pharmacology , Cells, Cultured , Chloride-Bicarbonate Antiporters/antagonists & inhibitors , Cyclooxygenase Inhibitors/pharmacology , Enterocytes/drug effects , Enzyme Inhibitors/pharmacology , Ileitis/genetics , Indoles/pharmacology , Lipoxygenase/metabolism , Lipoxygenase Inhibitors/pharmacology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Piroxicam/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL