Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 568
Filter
1.
Biol Pharm Bull ; 44(2): 275-278, 2021.
Article in English | MEDLINE | ID: mdl-33518681

ABSTRACT

α-Defensin 5 has a particularly broad antibacterial spectrum; it eliminates pathogenic microorganisms and regulates intestinal flora. Although Caco-2 cells are similar to small intestinal cells, it is unclear whether they secrete α-defensin 5. Therefore, we investigated whether Caco-2 cells secrete α-defensin 5 and determined the secretion mechanism using cells from three cell banks (ATCC, DSMZ, and RIKEN). The Caco-2 cell proliferation rate increased with the number of culture days, irrespective of cell bank origin. On the other hand, the alkaline phosphatase activity, which affects cell differentiation and the mRNA levels of several cytokines, such as interleukin 8 (IL-8), IL-6, IL-1ß, tumor necrosis factor-α (TNF-α), and IL-2, in the Caco-2 cells fluctuated with the number of culture days, and differed for each cell bank. α-Defensin 5 secretion was detected in all three cell bank Caco-2 cells; particularly, the ATCC Caco-2 cells grew linearly depending on the cell culture day as well as the levels of IL-8 and TNF-α mRNA. This suggested that α-defensin 5 secretion in the ATCC Caco-2 cells was associated with fluctuations in the mRNA levels of various cytokines, such as IL-8 and TNF-α. In conclusion, Caco-2 cells may be a simple model for screening health food components and drugs that affect α-defensin 5 secretion.


Subject(s)
Caco-2 Cells/metabolism , alpha-Defensins/metabolism , Biological Specimen Banks , Cell Proliferation , Cytokines/analysis , Cytokines/metabolism , Drug Evaluation, Preclinical/methods , Feasibility Studies , Humans , Reproducibility of Results , alpha-Defensins/analysis
2.
J Sci Food Agric ; 101(8): 3156-3164, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33211321

ABSTRACT

BACKGROUND: Rubus chingii Hu is a widely cultivated fruit in China and has declared multiple bioactivities including antioxidative activity. Ethyl carbamate (EC), mostly found in fermented food and alcoholic beverages, is a recognized human carcinogen, and researchers have proposed the correlation between oxidative stress and its toxicity. This study acquired the polysaccharide from R. chingii (RP) and explored its effect on EC-induced cytotoxicity using Caco-2 cells as the cell model. RESULTS: Results showed that RP exhibited protection against EC-induced toxicity by repairing redox imbalance as indicative of mitigated mitochondrial membrane potential collapse, attenuated reactive oxygen species overproduction, and impeded glutathione depletion. Moreover, the structural features of RP were characterized and revealed that it was mainly constituted by galacturonic acid and arabinose, with an average molecular weight of 7.039 × 105 g mol-1 . CONCLUSION: Overall, our results provided a new approach dealing with the toxicity caused by EC from the perspective of oxidative stress and described a new potential healthy value of R. chingii Hu, which could contribute to the development of a promising dietary supplement and functional food. © 2020 Society of Chemical Industry.


Subject(s)
Plant Extracts/pharmacology , Polysaccharides/pharmacology , Protective Agents/pharmacology , Rubus/chemistry , Urethane/toxicity , Antioxidants , Caco-2 Cells/drug effects , Caco-2 Cells/metabolism , Cell Survival/drug effects , Glutathione/metabolism , Humans , Oxidative Stress/drug effects , Plant Extracts/isolation & purification , Polysaccharides/isolation & purification , Protective Agents/isolation & purification , Reactive Oxygen Species/metabolism
3.
Mol Pharm ; 17(3): 757-768, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32011888

ABSTRACT

Salmon calcitonin (sCT) is a potent calcium-regulating peptide hormone and widely applied for the treatment of some bone diseases clinically. However, the therapeutic usefulness of sCT is hindered by the frequent injection required, owing to its short plasma half-life and therapeutic need for a high dose. Oral delivery is a popular modality of administration for patients because of its convenience to self-administration and high patient compliance, while orally administered sCT remains a great challenge currently due to the existence of multiple barriers in the gastrointestinal (GI) tract. Here, we introduced an orally targeted delivery system to increase the transport of sCT across the intestine through both the paracellular permeation route and the bile acid pathway. In this system, sCT-based glycol chitosan-taurocholic acid conjugate (GC-T)/dextran sulfate (DS) ternary nanocomplexes (NC-T) were produced by a flash nanocomplexation (FNC) process in a kinetically controlled mode. The optimized NC-T exhibited well-controlled properties with a uniform and sub-60 nm hydrodynamic diameter, high batch-to-batch reproducibility, good physical or chemical stability, as well as sustained drug release behaviors. The studies revealed that NC-T could effectively improve the intestinal uptake and permeability, owing to its surface functionalization with the taurocholic acid ligand. In the rat model, orally administered NC-T showed an obvious hypocalcemia effect and a relative oral bioavailability of 10.9%. An in vivo assay also demonstrated that NC-T induced no observable side effect after long-term oral administration. As a result, the orally targeted nanocomplex might be a promising candidate for improving the oral transport of therapeutic peptides.


Subject(s)
Calcitonin/administration & dosage , Calcium-Regulating Hormones and Agents/administration & dosage , Drug Delivery Systems/methods , Intestinal Absorption/drug effects , Nanocomposites/chemistry , Administration, Oral , Animals , Biological Availability , Biological Transport , Caco-2 Cells/drug effects , Caco-2 Cells/metabolism , Calcitonin/adverse effects , Calcitonin/blood , Calcitonin/pharmacokinetics , Calcium/blood , Calcium-Regulating Hormones and Agents/adverse effects , Calcium-Regulating Hormones and Agents/blood , Calcium-Regulating Hormones and Agents/pharmacokinetics , Chitosan/chemistry , Dextran Sulfate/chemistry , Drug Liberation , Drug Stability , Half-Life , Humans , Hypocalcemia/chemically induced , Injections, Subcutaneous , Male , Rats , Rats, Sprague-Dawley , Taurocholic Acid/chemistry
4.
Article in English | MEDLINE | ID: mdl-31481446

ABSTRACT

P-glycoprotein (ABCB1), an ATP-binding-cassette efflux transporter, limits intestinal absorption of its substrates and is a common site of drug-drug interactions (DDIs). ABCB1 has been suggested to interact with many antivirals used to treat HIV and/or chronic hepatitis C virus (HCV) infections. Using bidirectional transport experiments in Caco-2 cells and a recently established ex vivo model of accumulation in precision-cut intestinal slices (PCIS) prepared from rat ileum or human jejunum, we evaluated the potential of anti-HIV and anti-HCV antivirals to inhibit intestinal ABCB1. Lopinavir, ritonavir, saquinavir, atazanavir, maraviroc, ledipasvir, and daclatasvir inhibited the efflux of a model ABCB1 substrate, rhodamine 123 (RHD123), in Caco-2 cells and rat-derived PCIS. Lopinavir, ritonavir, saquinavir, and atazanavir also significantly inhibited RHD123 efflux in human-derived PCIS, while possible interindividual variability was observed in the inhibition of intestinal ABCB1 by maraviroc, ledipasvir, and daclatasvir. Abacavir, zidovudine, tenofovir disoproxil fumarate, etravirine, and rilpivirine did not inhibit intestinal ABCB1. In conclusion, using recently established ex vivo methods for measuring drug accumulation in rat- and human-derived PCIS, we have demonstrated that some antivirals have a high potential for DDIs on intestinal ABCB1. Our data help clarify the molecular mechanisms responsible for reported increases in the bioavailability of ABCB1 substrates, including antivirals and drugs prescribed to treat comorbidity. These results could help guide the selection of combination pharmacotherapies and/or suitable dosing schemes for patients infected with HIV and/or HCV.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Anti-HIV Agents/pharmacology , Antiviral Agents/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Aged , Animals , Atazanavir Sulfate/pharmacology , Benzimidazoles/pharmacology , Caco-2 Cells/drug effects , Caco-2 Cells/metabolism , Carbamates , Drug Interactions , Female , Fluorenes/pharmacology , HIV Infections/complications , HIV Infections/drug therapy , HIV Infections/virology , Hepatitis C/complications , Hepatitis C/drug therapy , Hepatitis C/virology , Humans , Imidazoles/pharmacology , Intestines/drug effects , Lopinavir/pharmacology , Male , Maraviroc/pharmacology , Middle Aged , Pyrrolidines , Rats , Rats, Wistar , Ritonavir/pharmacology , Saquinavir/pharmacology , Valine/analogs & derivatives , Zidovudine/pharmacology
5.
J Bioenerg Biomembr ; 51(6): 403-412, 2019 12.
Article in English | MEDLINE | ID: mdl-31845097

ABSTRACT

Candida albicans causes mucosal diseases and secretes farnesol, a quorum-sensing molecule, which plays a vital role in suppressing the yeast-to-mycelia switch. Farnesol can also regulate immune cell function. However, how farnesol interacts with the intestinal epithelium remains unknown. Herein, we identified that farnesol promotes intestinal barrier function, by promoting transepithelial electrical resistance, reducing paracellular flux, inducing the Zonula Occludens-1 Protein (ZO-1) and occludin expression. Moreover, the JAK/STAT3 signaling pathway was activated after farnesol treatment, and inhibition of STAT3 phosphorylation by stattic remarkably suppressed the expression level of ZO-1. Additionally, chromatin immunoprecipitation assay (Chip) revealed that farnesol facilitated the transcriptional activation of STAT3 to significantly enhance the expression of ZO-1. Taken together, our findings demonstrated that farnesol facilitated intestinal epithelial barrier transcriptional regulation via activating JAK/STAT3 signaling. The involved molecules may be potentially targeted for treatment of Candida albicans invasion.


Subject(s)
Caco-2 Cells/metabolism , Epithelial Cells/metabolism , Farnesol/therapeutic use , Intestinal Mucosa/metabolism , STAT3 Transcription Factor/metabolism , Tight Junctions/metabolism , Cell Differentiation , Farnesol/pharmacology , Humans , Signal Transduction , Transfection
6.
Microb Pathog ; 120: 79-84, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29715536

ABSTRACT

Listeria monocytogenes expresses various virulence factors enabling the invasion and multiplying in host cells, and together induces cytokines transcription. In order to explore the relationship between virulence factors of L. monocytogenes wild-type EGD-e and cellular response in human colonic epithelial cell line(Caco-2), we constructed mutant strains with in-frame deletions of critical virulence genes of inlA, inlB, hly, actA and virulence regulatory factor prfA from EGD-e, respectively. Compared with EGD-e, mutant strains showed significantly decreased invasion and apoptosis in Caco-2 cells. However, mutant strains were capable to evoke cytokines transcription of interleukin-8 (IL-8), mononuclear chemoattractant protein-1 (MCP-1), tumor necrosis factor-a (TNF-a), interleukin-1ß (IL-1ß), interleukin-6 (IL-6) and CXCL-2 production in Caco-2 cells. Interestingly, EGD-e Δhly-infected Caco-2 cells showed a significant decrease of IL-6, IL-8 and MCP-1 transcription compared with EGD-e at 1 h post-infection. Simultaneously, EGD-e ΔinlB-infected cells showed a decrease in IL-6 transcription, while EGD-e ΔactA-infected cells reflected a decrease in MCP-1 transcription. Virulence genes play a role in inflammatory transcription, but the interaction between pathogenic bacteria and the host cells predominates in inflammatory transcription. Overall, the data showed cellular response of Caco-2 cells infected with EGD-e mutant strains.


Subject(s)
Caco-2 Cells/drug effects , Caco-2 Cells/metabolism , Listeria monocytogenes/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Virulence Factors/adverse effects , Apoptosis/drug effects , Bacterial Proteins/adverse effects , Bacterial Toxins/adverse effects , Caco-2 Cells/immunology , Chemokine CCL2/metabolism , Chemokine CXCL2/metabolism , Cytokines/metabolism , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/physiology , Humans , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Listeria monocytogenes/pathogenicity , Membrane Proteins/adverse effects , Peptide Termination Factors/adverse effects , Tumor Necrosis Factor-alpha/metabolism , Virulence/genetics , Virulence Factors/genetics
7.
Pharm Res ; 35(4): 74, 2018 Feb 26.
Article in English | MEDLINE | ID: mdl-29484506

ABSTRACT

PURPOSE: To identify conditions allowing the use of cell-based models for studies of drug absorption during in vitro lipolysis of lipid-based formulations (LBFs). METHODS: Caco-2 was selected as the cell-based model system. Monolayer integrity was evaluated by measuring mannitol permeability after incubating Caco-2 cells in the presence of components available during lipolysis. Pure excipients and formulations representing the lipid formulation classification system (LFCS) were evaluated before and after digestion. Porcine mucin was evaluated for its capacity to protect the cell monolayer. RESULTS: Most undigested formulations were compatible with the cells (II-LC, IIIB-LC, and IV) although some needed mucin to protect against damaging effects (II-MC, IIIB-MC, I-LC, and IIIA-LC). The pancreatic extract commonly used in digestion studies was incompatible with the cells but the Caco-2 monolayers could withstand immobilized recombinant lipase. Upon digestion, long chain formulations caused more damage to Caco-2 cells than their undigested counterparts whereas medium chain formulations showed better tolerability after digestion. CONCLUSIONS: Most LBFs and components thereof (undigested and digested) are compatible with Caco-2 cells. Pancreatic enzyme is not tolerated by the cells but immobilized lipase can be used in combination with the cell monolayer. Mucin is beneficial for critical formulations and digestion products.


Subject(s)
Caco-2 Cells/drug effects , Drug Evaluation, Preclinical/methods , Drug Liberation , Lipolysis , Pharmaceutical Preparations/metabolism , Administration, Oral , Caco-2 Cells/metabolism , Enzymes, Immobilized/metabolism , Enzymes, Immobilized/toxicity , Excipients/chemistry , Fungal Proteins , Gastrointestinal Absorption , Humans , Lipase/metabolism , Lipase/toxicity , Lipids/chemistry , Mucins/metabolism , Pancrelipase/metabolism , Pancrelipase/toxicity , Recombinant Proteins/metabolism
8.
Cell Biol Int ; 42(1): 112-120, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28914468

ABSTRACT

Alpha-gliadin peptide 31-43 is considered to be the main peptide responsible for the innate immune response in celiac disease patients. Recent evidence indicates that peptide 31-43 rapidly enters cells and interacts with the early endocytic vesicular compartment. However, the mechanism of its uptake is not completely understood. Our aim is to characterize, isolate and identify possible cell surface proteins involved in peptide 31-43 internalization by Caco-2 cells. In this study, we used a chemical cross-linker to block peptide 31-43 on cell surface proteins, and pulled-down peptide-proteins complexes using antibodies raised against peptide 31-43. Through this experimental approach, we did not observe any specific complex between cell proteins and peptide 31-43 in Coomassie-stained denaturating gels or by Western blotting. We also found that type 2 transglutaminase was not necessary for peptide 31-43 internalization, even though it had a regulatory role in the process. Finally, we demonstrated that peptide 31-43 did not behave as a classical ligand, indeed the labeled peptide did not displace the unlabeled peptide in a competitive binding assay. On the basis of these findings and of previous evidence demonstrating that peptide 31-43 is able to interact with a membrane-like environment in vitro, we conclude that membrane composition and organization, rather than a specific receptor protein, may have a major role in peptide 31-43 internalization by cells.


Subject(s)
Endocytosis/physiology , Gliadin/metabolism , Antibodies/immunology , Caco-2 Cells/metabolism , Celiac Disease/immunology , Celiac Disease/physiopathology , Cell Count , GTP-Binding Proteins , Gliadin/toxicity , HEK293 Cells/metabolism , Humans , Immunity, Innate/immunology , Immunity, Innate/physiology , Intestinal Mucosa/metabolism , Peptide Fragments/metabolism , Peptides/metabolism , Protein Glutamine gamma Glutamyltransferase 2 , Receptors, Cell Surface/physiology , Transglutaminases
9.
Gastroenterology ; 150(1): 168-80, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26433161

ABSTRACT

BACKGROUND & AIMS: Enteric glial cells (EGCs) produce soluble mediators that regulate homeostasis and permeability of the intestinal epithelial barrier (IEB). We investigated the profile of polyunsaturated fatty acid (PUFA) metabolites produced by EGCs from rats and from patients with Crohn's disease (CD), compared with controls, along with the ability of one of these metabolites, 15-hydroxyeicosatetraenoic acid (15-HETE), to regulate the permeability of the IEB. METHODS: We isolated EGCs from male Sprague-Dawley rats, intestinal resections of 6 patients with CD, and uninflamed healthy areas of intestinal tissue from 6 patients who underwent surgery for colorectal cancer (controls). EGC-conditioned media was analyzed by high-sensitivity liquid-chromatography tandem mass spectrometry to determine PUFA signatures. We used immunostaining to identify 15-HETE-producing enzymes in EGCs and tissues. The effects of human EGCs and 15-HETE on permeability and transepithelial electrical resistance of the IEB were measured using Caco-2 cells; effects on signal transduction proteins were measured with immunoblots. Levels of proteins were reduced in Caco-2 cells using short-hairpin RNAs or proteins were inhibited pharmacologically. Rats were given intraperitoneal injections of 15-HETE or an inhibitor of 15-lipoxygenase (the enzyme that produces 15-HETE); colons were collected and permeability was measured. RESULTS: EGCs expressed 15-lipoxygenase-2 and produced high levels of 15-HETE, which increased IEB resistance and reduced IEB permeability. 15-HETE production was reduced in EGCs from patients with CD compared with controls. EGCs from patients with CD were unable to reduce the permeability of the IEB; the addition of 15-HETE restored permeability to levels of control tissues. Inhibiting 15-HETE production in rats increased the permeability of the IEB in colon tissues. We found that 15-HETE regulates IEB permeability by inhibiting an adenosine monophosphate-activated protein kinase and increasing expression of zonula occludens-1. CONCLUSIONS: Enteric glial cells from patients with CD have reduced production of 15-HETE, which controls IEB permeability by inhibiting adenosine monophosphate-activated protein kinase and increasing expression of zonula occludens-1.


Subject(s)
Cell Membrane Permeability/physiology , Crohn Disease/metabolism , Hydroxyeicosatetraenoic Acids/metabolism , Neuroglia/metabolism , Analysis of Variance , Animals , Blotting, Western , Caco-2 Cells/metabolism , Cells, Cultured , Disease Models, Animal , Humans , Immunohistochemistry , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Reference Values
10.
Cell Microbiol ; 18(7): 1024-40, 2016 07.
Article in English | MEDLINE | ID: mdl-26749168

ABSTRACT

Subtilase cytotoxin (SubAB) is mainly produced by locus of enterocyte effacement (LEE)-negative strains of Shiga-toxigenic Escherichia coli (STEC). SubAB cleaves an endoplasmic reticulum (ER) chaperone, BiP/Grp78, leading to induction of ER stress. This stress causes activation of ER stress sensor proteins and induction of caspase-dependent apoptosis. We found that SubAB induces stress granules (SG) in various cells. Aim of this study was to explore the mechanism by which SubAB induced SG formation. Here, we show that SubAB-induced SG formation is regulated by activation of double-stranded RNA-activated protein kinase (PKR)-like endoplasmic reticulum kinase (PERK). The culture supernatant of STEC O113:H21 dramatically induced SG in Caco2 cells, although subAB knockout STEC O113:H21 culture supernatant did not. Treatment with phorbol 12-myristate 13-acetate (PMA), a protein kinase C (PKC) activator, and lysosomal inhibitors, NH4 Cl and chloroquine, suppressed SubAB-induced SG formation, which was enhanced by PKC and PKD inhibitors. SubAB attenuated the level of PKD1 phosphorylation. Depletion of PKCδ and PKD1 by siRNA promoted SG formation in response to SubAB. Furthermore, death-associated protein 1 (DAP1) knockdown increased basal phospho-PKD1(S916) and suppressed SG formation by SubAB. However, SG formation by an ER stress inducer, Thapsigargin, was not inhibited in PMA-treated cells. Our findings show that SubAB-induced SG formation is regulated by the PERK/DAP1 signalling pathway, which may be modulated by PKCδ/PKD1, and different from the signal transduction pathway that results in Thapsigargin-induced SG formation.


Subject(s)
Cytoplasmic Granules/metabolism , Escherichia coli Proteins/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Subtilisins/metabolism , Apoptosis Regulatory Proteins/metabolism , Caco-2 Cells/drug effects , Caco-2 Cells/metabolism , Caco-2 Cells/microbiology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chloroquine/pharmacology , Culture Media, Conditioned/pharmacology , DNA Helicases , Endoplasmic Reticulum Chaperone BiP , Escherichia coli Proteins/genetics , Escherichia coli Proteins/pharmacology , Gene Knockout Techniques , HeLa Cells , Host-Pathogen Interactions , Humans , Poly-ADP-Ribose Binding Proteins , Protein Kinase C-delta/metabolism , RNA Helicases , RNA Recognition Motif Proteins , Shiga-Toxigenic Escherichia coli/pathogenicity , Signal Transduction/drug effects , Stress, Physiological/drug effects , Subtilisins/genetics , Subtilisins/pharmacology , eIF-2 Kinase/metabolism
11.
Biol Cell ; 108(1): 19-28, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26526116

ABSTRACT

BACKGROUND INFORMATION: Microvillus inclusion disease (MVID) is a genetic disorder affecting intestinal absorption. It is caused by mutations in MYO5B or syntaxin 3 (STX3) affecting apical membrane trafficking. Morphologically, MVID is characterised by a depletion of apical microvilli and the formation of microvillus inclusions inside the cells, suggesting a loss of polarity. To investigate this hypothesis, we examined the location of essential apical polarity determinants in five MVID patients. RESULTS: We found that the polarity determinants Cdc42, Par6B, PKCζ/ι and the structural proteins ezrin and phospho-ezrin were lost from the apical membrane and accumulated either in the cytoplasm or on the basal side of enterocytes in patients, which suggests an inversion of cell polarity. Moreover, microvilli-like structures were observed at the basal side as per electron microscopy analysis. We next performed Myo5B depletion in three dimensional grown human Caco2 cells forming cysts and found a direct link between the loss of Myo5B and the mislocalisation of the same apical proteins; furthermore, we observed that a majority of cysts displayed an inverted polarity phenotype as seen in some patients. Finally, we found that this loss of polarity was specific for MVID: tissue samples of patients with Myo5B-independent absorption disorders showed normal polarity but we identified Cdc42 as a potentially essential biomarker for trichohepatoenteric syndrome. CONCLUSION: Our findings indicate that the loss of Myo5B induces a strong loss of enterocyte polarity, potentially leading to polarity inversion. SIGNIFICANCE: Our results show that polarity determinants could be useful markers to help establishing a diagnosis in patients. Furthermore, they could be used to characterise other rare intestinal absorption diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Membrane/metabolism , Cell Polarity/physiology , Malabsorption Syndromes/metabolism , Microvilli/pathology , Mucolipidoses/metabolism , cdc42 GTP-Binding Protein/metabolism , Caco-2 Cells/metabolism , Enterocytes/metabolism , Humans , Malabsorption Syndromes/pathology , Microvilli/metabolism , Mucolipidoses/pathology , Mutation/genetics , Myosin Heavy Chains/metabolism , Myosin Type V/genetics , Myosin Type V/metabolism , Protein Transport/genetics , Protein Transport/physiology
12.
Mol Pharm ; 12(10): 3685-90, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26365521

ABSTRACT

In this work, we discuss leveraging the Biopharmaceutics Classification System (BCS) in the development of edivoxetine HCl, a selective norepinephrine reuptake inhibitor. First, the biopharmaceutical and in vivo data are presented and discussed. Solubility studies indicate that edivoxetine HCl meets the BCS "highly soluble" criteria. To determine permeability classifications, in vitro intestinal Caco-2 epithelial cell model with and without cyclosporin A (CsA), a common P-glycoprotein (P-gp) inhibitor, were conducted. Pharmacokinetic (PK) data obtained across phase 1 and 2 clinical studies where single and multiple doses range from the lowest to the highest strength are presented. Neither the Caco-2 nor the in vivo data on their own were sufficient to conclusively classify edivoxetine as highly permeable. However, collectively the data were utilized to support high permeability and consequently BCS1 classification of edivoxetine HCl. BCS1 classification was leveraged throughout development to assess the risk associated with not conducting relative bioavailability (RBA) studies and avoiding bioequivalence (BE) studies. Examples are presented where formulation changes were made between phase I (drug in capsule/drug in bottle formulations) and phase II (tablet) trials in addition to phase III (tablet) and commercial (smaller tablet) without having to conduct any in vivo comparability studies. For the first change, BCS was leveraged to avoid conducting a RBA study even before obtaining official BCS classification. For the later change, official BCS1 classification was relied upon to avoid conducting a BE study.


Subject(s)
Biopharmaceutics/classification , Drug Discovery/methods , Biological Availability , Biopharmaceutics/methods , Caco-2 Cells/metabolism , Capsules , Chemistry, Pharmaceutical/methods , Clinical Trials, Phase I as Topic/methods , Humans , Intestinal Absorption , Morpholines/administration & dosage , Morpholines/pharmacokinetics , Phenylethyl Alcohol/administration & dosage , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacokinetics , Solubility , Tablets
13.
Xenobiotica ; 45(8): 722-30, 2015.
Article in English | MEDLINE | ID: mdl-25761590

ABSTRACT

1. Absorption and metabolism of tiliroside (kaempferol 3-ß-D-(6"-p-coumaroyl)-glucopyranoside) and its related compounds kaempferol, kaempferol-3-glucoside and p-coumaric acid were investigated in the small intestinal Caco-2 cell model. Apparent permeation (Papp) was determined as 0.62 × 10(-6) cm/s, 3.1 × 10(-6) cm/s, 0 and 22.8 × 10(-6) cm/s, respectively. 2. Mechanistic study showed that the transportation of tiliroside, kaempferol-3-glucoside and p-coumaric acid in Caco-2 model were transporter(s) involved, while transportation of kaempferol was solely by passive diffusion mechanism. 3. Efflux transporters, multi-drug-resistance-associated protein-2 (MRP2), were shown to play a role in limiting the uptake of tiliroside. Inhibitors of MRP2, (MK571 and rifampicin) and co-incubation with kaempferol (10 µM), increased transfer from the apical to the basolateral side by three to five fold. 4. Metabolites of kaempferol-3-glucoside and p-coumaric acid were not detected in the current Caco-2 model, while tiliroside was metabolised to a limited extent, with two tiliroside mono-glucuronides identified; and kaempferol was metabolised to a higher extent, with three mono-glucuronides and two mono-sulfates identified. 5. In conclusion, tiliroside was metabolised and transported across Caco-2 cell membrane to a limited extent. Transportation could be increased by applying MRP2 inhibitors or co-incubation with kaempferol. It is proposed that tiliroside can be absorbed by human; future pharmacokinetics studies are warranted in order to determine the usefulness of tiliroside as a bioactive agent.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Coumaric Acids/metabolism , Flavonoids/metabolism , Intestine, Small/metabolism , Kaempferols/metabolism , Membrane Transport Proteins/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Biological Availability , Biological Transport , Caco-2 Cells/metabolism , Humans , Intestinal Absorption , Propionates , Time Factors
14.
Int J Mol Sci ; 16(9): 21555-74, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26370977

ABSTRACT

Anthocyanins from different plant sources have been shown to possess health beneficial effects against a number of chronic diseases. To obtain any influence in a specific tissue or organ, these bioactive compounds must be bioavailable, i.e., effectively absorbed from the gut into the circulation and transferred to the appropriate location within the body while still maintaining their bioactivity. One of the key factors affecting the bioavailability of anthocyanins is their transport through the gut epithelium. The Caco-2 cell line, a human intestinal epithelial cell model derived from a colon carcinoma, has been proven to be a good alternative to animal studies for predicting intestinal absorption of anthocyanins. Studies investigating anthocyanin absorption by Caco-2 cells report very low absorption of these compounds. However, the bioavailability of anthocyanins may be underestimated since the metabolites formed in the course of digestion could be responsible for the health benefits associated with anthocyanins. In this review, we critically discuss recent findings reported on the anthocyanin absorption and metabolism by human intestinal Caco-2 cells.


Subject(s)
Anthocyanins/metabolism , Caco-2 Cells/metabolism , Intestinal Absorption , Intestinal Mucosa/metabolism , Animals , Anthocyanins/chemistry , Biological Availability , Biological Transport , Humans
15.
Pharmazie ; 70(12): 798-803, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26817277

ABSTRACT

Caco2 cells are a human adenocarcinoma cell line that forms tight junctions and are widely used to examine bidirectional drug transport as well as P-glycoprotein mediated efflux. Unfortunately Caco2 cell lines can be very heterogeneous in nature. Our aim was to improve the Caco2 cell model for determination of P-glycoprotein mediated drug transport. Young passage Caco2 from ATCC had inadequate expression of P-glycoprotein, therefore three approaches were adopted to upregulate Caco2 P-glycoprotein expression to mimic that in vivo; a) incubation of mature Caco2 monolayer with rifampicin, b) prolonged exposure of Caco2 cells to vinblastine (generating the Caco2 VIN line), and c) splitting cells every 7 to 9 days until late passage numbers (over P80) were available. Upon development of the models, P-gp expression and activity was determined using western blotting and bidirectional transport studies of rhodamine123. All four models exhibited P-gp mediated efflux transport for rhodamine123. Incubation with rifampicin did not alter bidirectional transport compared to passage 44 cells. Increased passage number altered P-glycoprotein expression and the efflux ratio increased to 4.7 for passage 80 from 1.4 of passage 44. The highest basolateral to apical transport was observed for both passage 89 Caco2 and the Caco2 VIN model with an efflux ratio of 13 to 14. Western blot images confirmed the increased P-glycoprotein expression of late passage and Caco2 VIN. Caco2 cells are not ready for P-gp related research when first acquired from ATCC (Passage 18). Late passage Caco2 cell monolayers or Caco2 VIN models are needed to determine P-gp mediated efflux transport.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Caco-2 Cells/metabolism , Pharmaceutical Preparations/metabolism , Biological Transport, Active , Blotting, Western , Cell Lineage , Fluorescent Dyes , Humans , Models, Biological , Rhodamine 123
16.
Plant Foods Hum Nutr ; 70(1): 1-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25575485

ABSTRACT

Vitamin A deficiency (VAD) remains a public health problem in some regions of Brazil. Increased use of orange-fleshed sweet potato (OFSP) as a source of pro-vitamin A represents a potential strategy for prevention of VAD. We compared the pro-vitamin A content, vitamin A equivalency and bioaccessibility of ß-carotene (ßC) of two varieties of home cooked OFSP and two commercial sources of processed OFSP. Pro-vitamin A carotenoid content in home cooked, Beauregard variety of OFSP exceeded that in Amelia variety and commercial products for babies. All-trans-ßC was the most abundant carotenoid in raw, cooked and commercial OFSP. Boiling and frying OFSP generally decreased total ßC. A serving of 100 g FW Beauregard variety of cooked OFSP contained greater than 100% of the estimated average requirement (EAR) for children and women, and up to 92% EAR for lactating women. Although the efficiency of micellarization of all-trans-ßC during simulated digestion of OFSP was relatively low (4-8%) and significantly less than for cis-isomers, the quantities of trans-ßC incorporated into micelles from boiled Beauregard and fried Amelia varieties exceeded that in micelles generated by digesting commercial OFSP. The bioaccessibility of pro-vitamin A carotenoids in the micelle fraction of digested OFSP was confirmed with differentiated cultures of Caco-2 human intestinal cells. Continued development of OFSP such as the Amelia and Beauregard varieties that are rich in trans-ßC and dissemination of best practices for home cooking are encouraged to increase consumption of this food to decrease the risk of vitamin A deficiency in Brazil.


Subject(s)
Caco-2 Cells/drug effects , Cooking/methods , Ipomoea batatas/chemistry , Vitamin A/metabolism , beta Carotene/pharmacokinetics , Biological Availability , Caco-2 Cells/metabolism , Cell Line , Hot Temperature , Humans , In Vitro Techniques , Plant Roots/chemistry , beta Carotene/analysis
17.
Am J Physiol Gastrointest Liver Physiol ; 306(3): G218-28, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24309183

ABSTRACT

Barrier dysfunction is pivotal to the pathogenesis of inflammatory bowel diseases (IBD) and collagenous colitis. Glucocorticoids restore barrier function in Crohn's disease, but whether this reflects attenuated inflammation or an epithelial-specific action has not yet been addressed. Using filter-grown Caco-2 monolayers as an in vitro model of the intestinal epithelial barrier, we observed that glucocorticoids induced a time- and dose-dependent increase in transepithelial electrical resistance (TEER) in a glucocorticoid receptor-dependent manner without altering flux of larger solutes or changing principal tight junction architecture. This was accompanied by reduced paracellular cation flux, reduced expression of the pore-forming tight junction component claudin-2, and upregulation of the sealing tight junction protein claudin-4. In contrast, expression of occludin, claudin-1, -7, or -8 was not altered. Dexamethasone increased expression and activity of MAPK phosphatase-1 and inhibition of this phosphatase prevented the glucocorticoid-induced changes in TEER and claudin expression, whereas inhibiting p38 or MEK1/2 was not sufficient to replicate the glucocorticoid effects. Upon exposure to IFN-γ, TNF-α, or IL-1ß, TEERs declined in dexamethasone-treated cells but remained consistently higher than in cells not receiving glucocorticoids. Treatment with IFN/TNF resulted in an upregulation of claudin-2 that was significantly attenuated by dexamethasone, whereas increased claudin-2 expression upon IL-1ß stimulation was not affected by glucocorticoids. Taken together, barrier augmentation might represent a previously unrecognized mechanism of action, potentially contributing to the therapeutic efficacy of glucocorticoids in IBD and collagenous colitis.


Subject(s)
Claudins/metabolism , Dual Specificity Phosphatase 1/metabolism , Epithelial Cells/metabolism , Glucocorticoids/pharmacology , Tight Junctions/metabolism , Anti-Inflammatory Agents/pharmacology , Caco-2 Cells/drug effects , Caco-2 Cells/metabolism , Cells, Cultured , Dexamethasone/pharmacology , Epithelial Cells/drug effects , Glucocorticoids/genetics , Glucocorticoids/metabolism , Humans , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Occludin/metabolism , Tight Junctions/drug effects , Tumor Necrosis Factor-alpha/metabolism
18.
Pharm Res ; 31(9): 2287-96, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24590880

ABSTRACT

PURPOSE: Poly-L-arginine (PLA) enhances the paracellular permeability of the Caco-2 cell monolayer to hydrophilic macromolecules by disappearance of tight junction (TJ) proteins from cell-cell junctions. However, the mechanism of the disappearance of TJ proteins in response to PLA has been unclear. In this study, we investigated the mechanism of disappearance of TJ proteins from cell-cell junctions after the application of PLA to Caco-2 cell monolayers. METHODS: The membrane conductance (Gt), FITC-dextran (FD-4) permeability, and localization of TJ proteins were examined after the treatment of Caco-2 cell monolayers with PLA in the presence of various endocytosis inhibitors. In addition, the localization of endosome marker proteins was also observed. RESULTS: Clathrin-mediated endocytosis inhibitors suppressed the increase in Gt and Papp of FD-4 induced by PLA, and also significantly suppressed the disappearance of TJ proteins induced by PLA. Furthermore, occludin, one of the TJ proteins, colocalized with early endosome and recycling endosomes after the internalization of occludin induced by PLA, and then was recycled to the cell-cell junctions. CONCLUSION: PLA induced the transient internalization of TJ proteins in cell-cell junctions via clathrin-mediated endocytosis, subsequently increasing the permeability of the Caco-2 cell monolayer to FD-4 via a paracellular route.


Subject(s)
Caco-2 Cells/drug effects , Clathrin/metabolism , Endocytosis/drug effects , Peptides/pharmacology , Tight Junction Proteins/metabolism , Biological Transport/drug effects , Caco-2 Cells/metabolism , Humans , Permeability/drug effects
19.
Minerva Gastroenterol Dietol ; 60(1): 71-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24632769

ABSTRACT

AIM: In this work we evaluated the possibility for dietary supplement COLOSTRO NONI to be used as preventive and therapeutic agent in various diseases characterized by altered intestinal homeostasis with changes in the composition of the microbiota, alteration of the morphology and functionality, and also inflammation of the epithelium. METHODS: Cellular activity of COLOSTRO NONI has been tested in an in vitro model of intestinal epithelium based on Caco-2 cell line. We tested the ability of COLOSTRO NONI to stimulate cellular turnover evaluating cell growth rate with WST-1 proliferation assay. We also tested the ability of COLOSTRO NONI to increase the gene expression of Interleukin-8 (IL-8) with a Real Time PCR assay. IL-8 is a fundamental chemotactic factor involved in inflammatory phenomena and in the control of tissue homeostasis. RESULTS: COLOSTRO NONI is able to stimulate cell turnover in the proposed in vitro model and demonstrates active in increasing the gene expression of IL-8. Both aspects observed are fundamental for the establishment of mechanisms to repair tissue damage. CONCLUSION: Obtained results indicate that COLOSTRO NONI could find clinical application in treatment of gastrointestinal disorders characterized by impairment of proper intestinal permeability, in inflammatory bowel diseases, in dysenteric diseases, in gastritis and in forms of pathological alteration of the mucous layer as celiac disease and gluten sensitivity.


Subject(s)
Colostrum , Fruit , Interleukin-8/metabolism , Intestinal Mucosa/drug effects , Morinda , Phytotherapy , Plant Extracts/therapeutic use , Animals , Caco-2 Cells/metabolism , Cattle , Female , Gastrointestinal Diseases/drug therapy , Gene Expression/drug effects , Humans , In Vitro Techniques , Inflammation/metabolism , Inflammation/pathology , Interleukin-8/genetics , Phytotherapy/methods , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Polymerase Chain Reaction , Pregnancy
20.
J Proteome Res ; 12(4): 1604-18, 2013 Apr 05.
Article in English | MEDLINE | ID: mdl-23387933

ABSTRACT

Clostridium difficile is the major cause of intestinal infections in hospitals. The major virulence factors are toxin A (TcdA) and toxin B (TcdB), which belong to the group of clostridial glucosylating toxins (CGT) that inactivate small GTPases. After a 24 h incubation period with TcdA or a glucosyltransferase-deficient mutant TcdA (gdTcdA), quantitative changes in the proteome of colonic cells (Caco-2) were analyzed using high-resolution LC-MS/MS and the SILAC technique. The changes in abundance of more than 5100 proteins were quantified. Nearly 800 toxin-responsive proteins were identified that were involved in cell cycle, cell structure, and adhesion as well as metabolic processes. Several proteins localized to mitochondria or involved in lipid metabolism were consistently of higher abundance after TcdA treatment. All changes of protein abundance depended on the glucosyltransferase activity of TcdA. Glucosylation of the known targets of TcdA such as RhoA, RhoC, RhoG was detected by LC-MS/MS. In addition, an almost complete glucosylation of Rap1(A/B), Rap2(A/B/C) and a partial glucosylation of Ral(A/B) and (H/K/N)Ras were detected. The glucosylation pattern of TcdA was compared to that of other CGT like TcdB, the variant TcdB from C. difficile strain VPI 1470 (TcdBF), and lethal toxin from C. sordellii (TcsL).


Subject(s)
Bacterial Toxins/metabolism , Caco-2 Cells/metabolism , Enterotoxins/metabolism , Host-Pathogen Interactions , Proteins/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/pharmacology , Bacterial Toxins/genetics , Bacterial Toxins/pharmacology , Caco-2 Cells/drug effects , Clostridioides difficile/metabolism , Clostridioides difficile/pathogenicity , Enterotoxins/genetics , Enterotoxins/pharmacology , Glycosylation , Humans , Proteins/analysis , Proteomics/methods , Substrate Specificity , Tandem Mass Spectrometry , rap GTP-Binding Proteins , rap1 GTP-Binding Proteins , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , rhoC GTP-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL