Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 19.914
Filter
1.
Food Chem ; 462: 140975, 2025 Jan 01.
Article in English | MEDLINE | ID: mdl-39197240

ABSTRACT

This study isolated a novel peptide MMGGED with strong calcium-binding capacity from defatted walnut meal and synthesized a novel peptide­calcium chelate COS-MMGGED-Ca with high stability via glycation. Structural characterization and computer simulation identified binding sites, while in vitro digestion stability and calcium transport experiments explored the chelate's properties. Results showed that after glycation, COS-MMGGED bound Ca2+ with 88.75 ± 1.75 %, mainly via aspartic and glutamic acids. COS-MMGGED-Ca released Ca2+ steadily (60.27 %), with thermal denaturation temperature increased by 18 °C and 37 °C compared to MMGGED-Ca, indicating good processing performance. Furthermore, COS-MMGGED significantly enhanced Ca2+ transport across Caco-2 monolayers, 1.13-fold and 1.62-fold higher than CaCl2 and MMGGED, respectively, at 240 h. These findings prove glycation enhances structural properties, stability, calcium loading, and transport of peptide­calcium chelates, providing a scientific basis for developing novel efficient calcium supplements and high-value utilization of walnut meal.


Subject(s)
Calcium , Juglans , Peptides , Juglans/chemistry , Humans , Calcium/chemistry , Calcium/metabolism , Caco-2 Cells , Peptides/chemistry , Peptides/metabolism , Glycosylation , Calcium Chelating Agents/chemistry
2.
Food Chem ; 462: 141030, 2025 Jan 01.
Article in English | MEDLINE | ID: mdl-39241685

ABSTRACT

The human milk fat globule membrane (hMFGM) and Lactobacillus modulate the infant's gut and benefit health. Hence, the current study assesses the probiotic potential of Lactiplantibacillus plantarum (MRK3), Limosilactobacillus ferementum (MK1) isolated from infant feces, and its interaction with hMFGM during conditions mimicking infant digestive tract. Both strains showed high tolerance to gastrointestinal conditions, cell surface hydrophobicity, and strong anti-pathogen activity against Staphylococcus aureus. During digestion, hMFGM significantly exhibited xanthine oxidase activity, membrane roughness, and surface topography. In the presence of hMFGM, survival of MRK3 was higher than MK1, and electron microscopic observation revealed successful entrapment of MRK3 in the membrane matrix throughout digestion. Interestingly, probiotic-membrane matrix interaction showed significant synergy to alleviate oxidative stress and damage induced by cell-free supernatant of Escherichia coli in Caco-2 cells. Our results show that a probiotic-encapsulated membrane matrix potentially opens the functional infant formula development pathway.


Subject(s)
Glycolipids , Glycoproteins , Lipid Droplets , Milk, Human , Oxidative Stress , Probiotics , Humans , Probiotics/pharmacology , Probiotics/chemistry , Lipid Droplets/chemistry , Lipid Droplets/metabolism , Glycoproteins/chemistry , Glycoproteins/pharmacology , Glycoproteins/metabolism , Caco-2 Cells , Glycolipids/chemistry , Glycolipids/pharmacology , Glycolipids/metabolism , Oxidative Stress/drug effects , Milk, Human/chemistry , Infant , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Infant Formula/chemistry , Escherichia coli/drug effects , Escherichia coli/metabolism , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/metabolism
3.
Food Chem ; 462: 140953, 2025 Jan 01.
Article in English | MEDLINE | ID: mdl-39216374

ABSTRACT

The study examined the antihypertensive effect of peptides derived from pepsin-hydrolyzed corn gluten meal, namely KQLLGY and PPYPW, and their in silico gastrointestinal tract digested fragments, KQL and PPY, respectively. KQLLGY and PPYPW showed higher angiotensin I-converting enzyme (ACE)-inhibitory activity and lower ACE inhibition constant (Ki) values when compared to KQL and PPY. Only KQL showed a mild antihypertensive effect in spontaneously hypertensive rats with -7.83 and - 5.71 mmHg systolic and diastolic blood pressure values, respectively, after 8 h oral administration. During passage through Caco-2 cells, KQL was further degraded to QL, which had reduced ACE inhibitory activity. In addition, molecular dynamics revealed that the QL-ACE complex was less stable compared to the KQL-ACE. This study reveals that structural transformation during peptide permeation plays a vital role in attenuating antihypertensive effect of the ACE inhibitor peptide.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors , Antihypertensive Agents , Peptidyl-Dipeptidase A , Zea mays , Animals , Humans , Male , Rats , Angiotensin-Converting Enzyme Inhibitors/chemistry , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/metabolism , Antihypertensive Agents/chemistry , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Caco-2 Cells , Digestion/drug effects , Gastrointestinal Tract/metabolism , Glutens/chemistry , Glutens/metabolism , Hydrolysis , Hypertension/metabolism , Hypertension/drug therapy , Hypertension/physiopathology , Peptides/chemistry , Peptides/pharmacology , Peptidyl-Dipeptidase A/chemistry , Peptidyl-Dipeptidase A/metabolism , Protein Hydrolysates/chemistry , Protein Hydrolysates/pharmacology , Rats, Inbred SHR , Zea mays/chemistry , Zea mays/metabolism
4.
Yale J Biol Med ; 97(3): 297-308, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39351327

ABSTRACT

The gut microbiota is a very important factor in the state of health of an individual, its alteration implies a situation of "dysbiosis," which can be connected to functional gastrointestinal disorders and pathological conditions, such as Inflammatory Bowel Disease (IBD), Irritable Bowel Syndrome (IBS), Ulcerative Colitis (UC) and Crohn's Disease (CD), and Colorectal Cancer (CRC). In this work, we studied the effect of a food supplement called ENTERO-AD containing a mix of probiotics (Lactobacillus acidophilus LA1, L. reuteri LR92, Bifidobacterium breve Bbr8), Matricaria Chamomilla, and B group vitamins (B1, B2, B6) on intestinal inflammation. The in vitro model used for the study is the Caco-2 cell, a culture derived from human intestinal adenocarcinoma; the inflammatory condition was achieved with treatment with Lipopolysaccharide (LPS) and the association between Tumor necrosis factor α/Interferon γ (TNF-α/IFN-γ) [1,2]. The effect of ENTERO-AD was evaluated by cell viability, measures of Transepithelial Electrical Resistance (TEER), paracellular permeability, and immunofluorescence. Results of the study have shown that ENTERO-AD has a favorable effect on Caco-2 cells in inflammatory conditions. It improves the integrity of Occludin and Zonula Occludens-1 (ZO-1) proteins, leading to an improvement in terms of TEER values and a reduction of paracellular permeability. This evidence underlines the protective effect of ENTERO-AD and its components in intestinal inflammation.


Subject(s)
Dietary Supplements , Intestinal Mucosa , Plant Extracts , Probiotics , Humans , Probiotics/pharmacology , Probiotics/administration & dosage , Caco-2 Cells , Plant Extracts/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Vitamins/pharmacology , Vitamins/administration & dosage , Cell Survival/drug effects , Inflammation/pathology , Inflammatory Bowel Diseases
5.
Carbohydr Polym ; 344: 122518, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39218543

ABSTRACT

A wide range of articles describe the role of different probiotics in the prevention or treatment of various diseases. However, currently, the focus is shifting from whole microorganisms to their easier-to-define components that can confer similar or stronger benefits on the host. Here, we aimed to describe polysaccharide B.PAT, which is a surface antigen isolated from Bifidobacterium animalis ssp. animalis CCDM 218 and to understand the relationship between its structure and function. For this reason, we determined its glycerol phosphate-substituted structure, which consists of glucose, galactose, and rhamnose residues creating the following repeating unit: To fully understand the role of glycerol phosphate substitution on the B.PAT function, we prepared the dephosphorylated counterpart (B.MAT) and tested their immunomodulatory properties. The results showed that the loss of glycerol phosphate increased the production of IL-6, IL-10, IL-12, and TNF-α in bone marrow dendritic cells alone and after treatment with Lacticaseibacillus rhamnosus GG. Further studies indicated that dephosphorylation can enhance B.PAT properties to suppress IL-1ß-induced inflammatory response in Caco-2 and HT-29 cells. Thus, we suggest that further investigation of B.PAT and B.MAT may reveal distinct functionalities that can be exploited in the treatment of various diseases and may constitute an alternative to probiotics.


Subject(s)
Bifidobacterium animalis , Humans , Phosphorylation/drug effects , Bifidobacterium animalis/chemistry , Animals , Caco-2 Cells , Polysaccharides, Bacterial/pharmacology , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/isolation & purification , HT29 Cells , Probiotics/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mice , Immunologic Factors/pharmacology , Immunologic Factors/chemistry , Immunologic Factors/isolation & purification , Cytokines/metabolism , Lacticaseibacillus rhamnosus/chemistry
6.
Carbohydr Polym ; 346: 122639, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39245530

ABSTRACT

Molecular weight (Mw) of ligand-mediated nanocarriers plays a pivotal role in their architecture and properties. In this study, self-assembled ovalbumin (OVA)-loaded nanoparticles were meticulously engineered by starch polyelectrolytes with different Mw. Results unveiled that, tailoring Mw of GRGDS pentapeptides-grafted carboxymethyl starch (G-CMS) displayed strong binding-affinity and transport efficiency through microfold cells (M cells) pathway in the simulated intestinal epithelial cell monolayer in which M cells were randomly located in the Caco-2 cells monolayer. Notably, nanoparticles assembled from G-CMS with relatively higher Mw exhibited more compact structures due to the stronger interactions between layers compared to that with relatively lower Mw, which rendered remarkably stable and only 19.01 % in vitro OVA leakage under conditions of the upper gastrointestinal tract. Subsequently, more intact nanoparticles reached M cells after in vitro digestion and exhibited higher transport efficiency through the M cells pathways (apparent permeability: 9.38 × 10-5 cm/s) than Caco-2 cells, attributing to specific- and non-specific binding affinity towards M cells. Therefore, optimal Mw tailoring of starch polyelectrolytes can mediate the molecular interactions among their assembled layers and the interactions with M cells to balance the structural compactness, release and transport efficacy of nanoparticles, holding promise for advancing M cells-targeting oral delivery technologies.


Subject(s)
Drug Carriers , Molecular Weight , Nanoparticles , Starch , Humans , Starch/chemistry , Starch/analogs & derivatives , Starch/metabolism , Caco-2 Cells , Nanoparticles/chemistry , Drug Carriers/chemistry , Ovalbumin/chemistry , Ovalbumin/metabolism , Drug Liberation , Biological Transport , M Cells
7.
Carbohydr Polym ; 346: 122647, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39245532

ABSTRACT

Incorporating 5-aminosalicylic acid (5-ASA) into a colon-specific carrier is crucial for treating inflammatory bowel diseases (IBD), as it enhances therapeutic efficacy, targets the affected regions directly, and minimizes side effects. This study evaluated the impact of incorporating cellulose nanofibers (CNF) on the in vitro and in vivo biological performance of retrograded starch/pectin (RS/P) microparticles (MPs) containing 5-ASA. Using Fourier Transform Infrared (FTIR) Spectroscopy, shifts in the spectra of retrograded samples containing CNF were observed with increasing CNF proportions, suggesting the establishment of new supramolecular interactions. Liquid absorption exhibited pH-dependent behaviors, with reduced absorption in simulated gastric fluid (∼269 %) and increased absorption in simulated colonic fluid (∼662 %). Increasing CNF concentrations enhanced mucoadhesion in porcine colonic sections, with a maximum force of 3.4 N at 50 % CNF. Caco-2 cell viability tests showed biocompatibility across all tested concentrations (0.0625-2.0000 mg/mL). Evaluation of intestinal permeability in Caco-2 cell monolayers demonstrated up to a tenfold increase in 5-ASA permeation, ranging from 29 % to 48 %. An in vivo study using Galleria mellonella larvae, with inflammation induced by LPS, showed reduction of inflammation. Given the scalability of spray-drying, these findings suggest the potential of CNF-incorporated RS/P microparticles for targeted 5-ASA delivery in IBD.


Subject(s)
Inflammatory Bowel Diseases , Mesalamine , Nanofibers , Pectins , Starch , Mesalamine/chemistry , Mesalamine/pharmacology , Mesalamine/administration & dosage , Animals , Caco-2 Cells , Humans , Inflammatory Bowel Diseases/drug therapy , Nanofibers/chemistry , Nanofibers/toxicity , Swine , Pectins/chemistry , Starch/chemistry , Drug Carriers/chemistry , Drug Delivery Systems , Cell Survival/drug effects , Colon/drug effects , Colon/metabolism , Colon/pathology , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage
8.
Pathol Res Pract ; 262: 155520, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39217771

ABSTRACT

Carcinoma of the colon and rectum, also known as colorectal cancer, ranks as the third most frequently diagnosed malignancy globally. Sorafenib exhibits broad-spectrum antitumor activity against Raf, VEGF, and PDGF pathways in hepatocellular, thyroid, and renal cancers, but faces resistance in colorectal malignancies. 6-Shogaol, a prominent natural compound found in Zingiberaceae, exhibits antioxidant, anti-inflammatory, anticancer, and antiemetic properties. We investigated the influence of 6-shogaol on sorafenib's cytotoxic profile against colorectal cancer cell lines (HT-29, HCT-116, CaCo-2, and LS174T) through its effects on cellular accumulation and metabolism. Cytotoxicity was assessed using the sulpharodamine B assay, caspase-3 and c-PARP cleavage, cell cycle distribution analysis, and P-gp efflux activity. 6-Shogoal showed considerable cytotoxicity with decreased IC50 in colorectal cancer cell lines. Combining sorafenib and 6-shogaol increased c-PARP and pro-caspase-3 concentrations in HCT-116 cells compared to sorafenib alone. In combination, pro-caspase-3 concentrations were decreased in CaCo-2 cells compared to alone. Sorafenib combinations with 6-shogaol showed a significant drop in cell cycle distribution from 16.96±1.10 % to 9.16±1.85 %, respectively. At 100 µM, sorafenib and 6-shogaol showed potent and significant activity with intra-cellular rhodamine concentration on P-gp efflux activity in CRC cell lines. In conclusion, 6-shogaol substantially improved the cytotoxic profile of sorafenib by affecting its cellular uptake and metabolism. Future research should focus on dosage optimization and formulation and evaluate the efficacy and safety of the combination in animal models with colorectal cancer.


Subject(s)
Catechols , Colorectal Neoplasms , Sorafenib , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Sorafenib/pharmacology , Catechols/pharmacology , Antineoplastic Agents/pharmacology , Drug Synergism , Apoptosis/drug effects , Cell Line, Tumor , Caco-2 Cells , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
9.
Biol Pharm Bull ; 47(9): 1477-1483, 2024.
Article in English | MEDLINE | ID: mdl-39231687

ABSTRACT

Salmon milt extract (SME) is rich in nucleotides, especially deoxyribonucleoside monophosphates (dNMPs), which has the potential to exert anti-obesity effects. Sodium-dependent glucose transporter 1 (SGLT1) and glucose transporter 2 (GLUT2) are responsible for absorbing sugar from the small intestine. The purpose of this study was to examine the effects of SME on the functions of SGLT1 and GLUT2 and elucidate the mechanisms underlying the inhibition of glucose absorption by SME. We investigated the effect of SME on the expression and function of intestinal glucose transporters, using differentiated Caco-2 cells. SME treatment decreased the expression SGLT1 and GLUT2 mRNA and protein in Caco-2 cells. [14C]-Labelled methyl-α-D-glucopyranoside and [3H]-labelled 2-deoxy-D-glucose (DG) uptake into Caco-2 cells was significantly reduced by SME treatment. Similarly, the dNMP mixture containing the four mononucleotides 2'-deoxyadenosine 5'-monophosphate (dAMP), 2'-deoxyguanosine 5'-monophosphate (dGMP), 2'-deoxycytidine 5'-monophosphate (dCMP), and 2'-deoxythymidine 5'-monophosphate (dTMP) decreased SGLT1 and GLUT2 expression. dNMP mixture-induced reduction in the mRNA expression of these transporters was suppressed when exposed to the mixture without dTMP. Furthermore, dNMP mixture-induced alterations in the expression of hepatocyte nuclear factor (HNF)-1α and HNF1ß, which have been characterized as modulators of both transporters also showed a similar trend. dTMP treatment alone decreased GLUT2 expression, resulting in reduced [3H] DG uptake by Caco-2 cells. SME decreased the expression of HNF1α, HNF1ß, and its targets SGLT1 and GLUT2, resulting in reduced glucose uptake by Caco-2 cells. In addition, our results revealed that dTMP plays an important role in suppressing the expression of intestinal glucose transporters.


Subject(s)
Down-Regulation , Glucose Transporter Type 2 , Glucose , Sodium-Glucose Transporter 1 , Humans , Caco-2 Cells , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 1/genetics , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 2/genetics , Glucose/metabolism , Down-Regulation/drug effects , Animals , Salmon , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 1-alpha/genetics
10.
AAPS PharmSciTech ; 25(7): 206, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39237659

ABSTRACT

Intestinal lymphatic transport offers an alternative and effective way to deliver drugs, such as avoiding first-pass metabolism, enhancing oral bioavailability, and facilitating the treatment of targeted lymphoid-related diseases. However, the clinical use of luteolin (LUT) is limited by its poor water solubility and low bioavailability, and enhancing lymphatic transport by nanoemulsion may be an efficient way to enhance its oral bioavailability. The objective of this work is to prepare the luteolin nanoemulsions (LUT NEs), optimized its preparation parameters by using Box-Behnken design optimization (BBD) and evaluated it in vitro and in vivo. An Caco-2 / Raji B cell co-incubation monolayer model was established to simulate the M-cell pathway, and the differences in the transmembrane transport of LUT and NEs were compared. Cycloheximide (CHX) was utilized to establish rat chylomicron (CM) blocking model, and for investigating the influence of pharmacokinetic parameters in rats thereafter. The results showed that LUT NEs have good stability, the particle sizes were about 23.87 ± 0.57 nm. Compared with LUT suspension, The Papp of LUT NEs was enhanced for 3.5-folds, the oral bioavailability was increased by about 2.97-folds. In addition, after binding with chylomicron, the oral bioavailability of LUT NEs was decreased for about 30% (AUC 0-∞ (µg/L*h): 5.356 ± 1.144 vs 3.753 ± 0.188). These results demonstrated that NEs could enhance the oral absorption of luteolin via lymphatic transport routes.


Subject(s)
Biological Availability , Emulsions , Luteolin , Nanoparticles , Particle Size , Rats, Sprague-Dawley , Luteolin/pharmacokinetics , Luteolin/administration & dosage , Luteolin/chemistry , Animals , Rats , Humans , Caco-2 Cells , Administration, Oral , Male , Nanoparticles/chemistry , Solubility , Intestinal Absorption/physiology , Chylomicrons/metabolism , Biological Transport/physiology , Lymphatic System/metabolism
11.
BMC Infect Dis ; 24(1): 905, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223499

ABSTRACT

BACKGROUND: Cholera is a diarrheal disease recognized for being caused by toxin-producing Vibrio (V.) cholerae. This study aims to assess the vibriocidal and immunomodulatory properties of derived cell-free supernatants (CFSs) of Bifidobacterium (B.) bifidum and Lactobacillus (L.) acidophilus encapsulated in chitosan nanoparticles (CFSb-CsNPs and CFSa-CsNPs) against clinical multi-drug resistance (MDR) isolates of V. cholerae O1 El Tor. METHODS: We synthesized CFSb-CsNPs and CFSa-CsNPs using the ionic gelation technique. The newly nanostructures were characterized for size, surface zeta potential, morphology, encapsulation efficacy (EE), stability in different pH values and temperatures, release profile, and in vitro cytotoxicity. The antimicrobial and antibiofilm effects of the obtained nanocomposites on clinical MDR isolates (N = 5) of V. cholerae E1 Tor O1 were investigated by microbroth dilution assay and crystal violet staining, respectively. We conducted quantitative real-time PCR (qRT-PCR) to analyze the relative gene expressions of Bap, Rbmc, CTXAB, and TCP in response to CFSb-CsNPs and CFSa-CsNPs. Additionally, the immunomodulatory effects of formulated structures on the expression of TLR2 and TLR4 genes in human colorectal adenocarcinoma cells (Caco-2) were studied. RESULTS: Nano-characterization analyses indicated that CFSb-CsNPs and CFSa-CsNPs exhibit spherical shapes under scanning electron microscopy (SEM) imaging, with mean diameters of 98.16 ± 0.763 nm and 83.90 ± 0.854 nm, respectively. Both types of nanoparticles possess positive surface charges. The EE% of CFSb-CsNPs was 77 ± 4.28%, whereas that of CFSa-CsNPs was 62.5 ± 7.33%. Chitosan (Cs) encapsulation leads to increased stability of CFSs in simulated pH conditions of the gastrointestinal tract in which the release rates for CFSb-CsNPs and CFSa-CsNPs were reached at 58.00 ± 1.24% and 55.01 ± 1.73%, respectively at pH = 7.4. The synergistic vibriocidal effects observed from the co-administration of both CFSb-CsNPs and CFSa-CsNPs, as evidenced by a fractional inhibitory concentration (FIC) index of 0.57, resulting in a significantly lower MIC of 2.5 ± 0.05 mg/mL (p < 0.0001) compare to individual administration. The combined antibacterial effect of CFSb-CsNPs and CFSa-CsNPs on Bap (0.14 ± 0.05), Rbmc (0.24 ± 0.01), CTXAB (0.30 ± 0.09), and TCP (0.38 ± 0.01) gene expression was significant (p < 0.001). Furthermore, co-administration of CFSb-CsNPs and CFSa-CsNPs also demonstrated the potency of suppressing TLR 2/4 (0.20 ± 0.01 and 0.12 ± 0.02, respectively) gene expression (p = 0.0019) and reduced Caco-2 cells attached bacteria to 526,000 ± 51,46 colony-forming units/mL (11.19%) (p < 0.0001). CONCLUSION: Our study revealed that encapsulating CFSs within CsNPs enhances their vibriocidal activity by improving stability and enabling a controlled release mechanism at the site of interaction between the host and bacteria. Additionally, the simultaneous use of CFSb-CsNPs and CFSa-CsNPs exhibited superior vibriocidal potency against MDR V. cholerae O1 El Tor strains, indicating these combinations as a potential new approach against MDR bacteria.


Subject(s)
Anti-Bacterial Agents , Bifidobacterium bifidum , Chitosan , Lactobacillus acidophilus , Nanoparticles , Vibrio cholerae O1 , Chitosan/chemistry , Chitosan/pharmacology , Lactobacillus acidophilus/drug effects , Vibrio cholerae O1/drug effects , Humans , Nanoparticles/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Bifidobacterium bifidum/physiology , Drug Resistance, Multiple, Bacterial , Probiotics/pharmacology , Probiotics/administration & dosage , Biofilms/drug effects , Microbial Sensitivity Tests , Caco-2 Cells
12.
Eur J Pharm Biopharm ; 203: 114480, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39222674

ABSTRACT

Efficient telmisartan delivery for hypertension management requires the incorporation of meglumine and/or sodium hydroxide as an alkalizer in the formulation. Long-term use of powerful alkalis with formulation as part of chronic therapy can cause metabolic alkalosis, ulcers, diarrhea, and body pain. Here, we aimed to design a telmisartan formulation without alkalizers. Telmisartan properties were tailor-made by microfluidizer-based physical modification. After microfluidization, telmisartan nanosuspension was lyophilized to obtain telmisartan premix powder. The optimized telmisartan nanosuspension had an average particle size of 579.85 ± 32.14 nm. The lyophilized premix was characterized by FT-IR, DSC, and PXRD analysis to ensure its physicochemical characteristics. The solubility analysis of premix showed 2.2 times, 2.3 times, and 6 times solubility improvement in 0.1 N HCl, phosphate buffer pH 7.5, and pH 6.8 compared to pure telmisartan. A 3D in-vitro Caco-2 model was developed to compare apparent permeability of API and powder premix. It showed that the powder premix was more permeable than pure API. The tablet formulation prepared from the telmisartan premix showed a dissolution profile comparable to that of the marketed formulation. The technique present herein can be used as a platform technology for solubility and permeability improvement of similar classes of molecules.


Subject(s)
Particle Size , Permeability , Solubility , Telmisartan , Telmisartan/administration & dosage , Telmisartan/pharmacokinetics , Telmisartan/chemistry , Humans , Caco-2 Cells , Drug Compounding/methods , Intestinal Absorption/drug effects , Powders/chemistry , Hydrogen-Ion Concentration , Nanoparticles/chemistry , Chemistry, Pharmaceutical/methods , Drug Liberation , Intestinal Barrier Function
13.
Nanotechnology ; 35(47)2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39240071

ABSTRACT

Inflammation involving adipose macrophages is an important inducer of obesity. Regulating macrophages polarization and improving the inflammatory microenvironment of adipose tissue is a new strategy for the treatment of obesity. An amphiphilic chondroitin sulfate phenylborate derivative (CS-PBE) was obtained by modifying the main chain of chondroitin sulfate with the hydrophobic small molecule phenylborate. Using CS-PBE self-assembly, macrophage targeting, reactive oxygen species (ROS) release and celastrol (CLT) encapsulation were achieved. The cytotoxicity, cellular uptake, internalization pathways and transmembrane transport efficiency of CS-PBE micelles were studied in Caco-2 and RAW264.7 cells. Hemolysis and organotoxicity tests were performed to assess the safety of the platform, while its therapeutic efficacy was investigated in high-fat diet-induced obese mice. Multifunctional micelles with macrophage targeting and ROS clearance capabilities were developed to improve the efficacy of CLT in treating obesity.In vitrostudies indicated that CS-PBE micelles had better ability to target M1 macrophages, better protective effects on mitochondrial function, better ability to reduce the number of LPS-stimulated M1 macrophages, better ability to reduce the number of M2 macrophages, and better ability to scavenge ROS in inflammatory macrophages.In vivostudies have shown that CS-PBE micelles improve inflammation and significantly reduce toxicity of CLT in the treatment of obesity. In summary, CS-PBE micelles could significantly improve the ability to target inflammatory macrophages and scavenge ROS in adipose tissue to alleviate inflammation, suggesting that CS-PBE micelles are a highly promising approach for the treatment of obesity.


Subject(s)
Macrophages , Micelles , Mitochondria , Obesity , Reactive Oxygen Species , Animals , Reactive Oxygen Species/metabolism , Mice , Obesity/drug therapy , Obesity/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Humans , Macrophages/drug effects , Macrophages/metabolism , RAW 264.7 Cells , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/pharmacology , Caco-2 Cells , Pentacyclic Triterpenes/pharmacology , Pentacyclic Triterpenes/chemistry , Mice, Inbred C57BL , Male , Diet, High-Fat/adverse effects , Triterpenes/pharmacology , Triterpenes/chemistry
14.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(8): 1545-1552, 2024 Aug 20.
Article in Chinese | MEDLINE | ID: mdl-39276050

ABSTRACT

OBJECTIVE: To investigate the mechanism by which swertiamarin (STM) ameliorates CD-like colitis in mice. METHODS: A Caco-2 cell model of TNF-α-stimulated apoptosis was established and divided into three groups: Con, TNF-α and STM, and the effects of STM on apoptosis and barrier function were assessed by Tunel staining, western blotting, immunofluorescence, and transepithelial electric resistance (TEER). A mouse model of 2, 4, 6-trinitrobenzenesulfonic acid (TNBS) -induced CD-like colitis was established to assess the effects of STM on colitis, intestinal barrier function and epithelial cell apoptosis. The regulatory role of the PI3K/AKT pathway in STM-induced resistance to intestinal epithelial cell apoptosis was investigated in both the cell model and mouse models. RESULTS: TUNEL staining showed that in Caco-2 cells with TNF-α stimulation, STM treatment significantly reduced the percentage of TUNEL-stained cells (P<0.05). STM obviously reduced TNF-α-induced enhancement of cleaved-caspase 3 and Bax expressions (P<0.05), increased Bcl-2 expression (P<0.05), protected intestinal barrier integrity and function by restoring transepithelial electrical resistance (TEER) of the cells, promoted normal localization and expressions of the tight junction proteins (ZO1 and claudin 1) (P<0.05), and inhibited the expression of pro-inflammatory factors (IL-6 and CCL3) (P<0.05) in TNF-α-stimulated Caco-2 cells. In the mouse models, STM significantly alleviated TNBS-induced CD-like colitis and intestinal barrier dysfunction (P<0.05) as shown by improved weight loss, lowered Disease Activity Index (DAI) score and inflammation score, reduction of IL-6 and CCL3 release, and restoration of intestinal barrier permeability, colonic TEER, bacterial translocation, and localization and expressions of the tight junction proteins. Mechanistically, STM inhibited the expressions of p-PI3K and p-AKT in both the cell model and mouse model(P<0.05), and treatment with 740Y-P (a PI3K/AKT pathway activator) significantly attenuated the inhibitory effect of STM on TNF-α-induced apoptosis in Caco-2 cells (P<0.05). CONCLUSION: STM inhibits intestinal epithelial cell apoptosis at least in part by suppressing activation of the PI3K/AKT pathway to ameliorate intestinal barrier dysfunction and colitis in mice.


Subject(s)
Apoptosis , Colitis , Epithelial Cells , Trinitrobenzenesulfonic Acid , Tumor Necrosis Factor-alpha , Animals , Mice , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Apoptosis/drug effects , Humans , Caco-2 Cells , Tumor Necrosis Factor-alpha/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Disease Models, Animal , Proto-Oncogene Proteins c-akt/metabolism , Iridoid Glucosides/pharmacology , Signal Transduction/drug effects , Caspase 3/metabolism
15.
Oncol Rep ; 52(5)2024 Nov.
Article in English | MEDLINE | ID: mdl-39329268

ABSTRACT

Accumulating evidence indicates that the dysregulation of microRNAs (miRNAs or miRs), is associated with human malignancies and suggests a casual role of miRNAs in tumor initiation and progression. Even though it has been discovered that a number of miRNAs play significant parts in the development of colorectal cancer (CRC), it is crucial to comprehend the regulatory functions that other miRNAs play in CRC. Based on GSE183437 and GSE156719 microarray data that were obtained from Gene Expression Omnibus database, candidate miRNAs were researched. The oncogenic effects of miR­25­3p in different malignancies have led to its selection for additional investigation in the present study. The expression of miR­25­3p was verified by reverse transcription­quantitative PCR, and its correlation with clinicopathological characteristics in patients with CRC was then investigated. In vitro assays were conducted to investigate the influence of miR­25­3p on the proliferative and apoptotic behaviors of HCT116 and Caco­2 cells. The present data revealed that miR­25­3p exhibited one of the most significant upregulations in CRC tissues and cell lines. The expression levels of miR­25­3p were found to be intimately correlated with tumor size, distant metastasis, tumor­node­metastasis stage, and shorter overall survival rate. In terms of functionality, the downregulation of miR­25­3p led to the inhibition of cellular proliferation and the enhancement of apoptosis in both HCT116 and Caco­2 cell lines. The critical tumor suppressor F­box and WD repeat containing domain 7 (FBXW7) was identified as a direct molecular target for miR­25­3p, with an inverse relationship observed between the two in neoplastic tissues. Subsequent studies demonstrated that the tumor suppressive effects of miR­25­3p inhibitor were effectively negated by the silencing of FBXW7. Moreover, the ability of FBXW7 to inhibit the expression of several oncogenes was deemed essential for countering the anticancer effects mediated by miR­25­3p downregulation. These findings posit miR­25­3p as a promising therapeutic target and prognostic indicator for CRC.


Subject(s)
Apoptosis , Cell Proliferation , Colorectal Neoplasms , F-Box-WD Repeat-Containing Protein 7 , Gene Expression Regulation, Neoplastic , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , F-Box-WD Repeat-Containing Protein 7/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , Male , Female , Cell Proliferation/genetics , Middle Aged , HCT116 Cells , Apoptosis/genetics , Caco-2 Cells , Aged , Prognosis , Adult , Carcinogenesis/genetics
16.
Nutrients ; 16(18)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39339664

ABSTRACT

Human breast milk (HBM) is the "gold standard" for infant nutrition. When breast milk is insufficient or unavailable, infant milk formula (IMF) can provide a safe and nutritious alternative. However, IMFs differ considerably from HBM in composition and health function. We compared the digestibility and potential health functions of IMF containing low cream (LC-) or high cream (HC-) with pooled HBM. After simulated infant digestion of these samples, the bioavailability of key nutrients and immunomodulatory activities were determined via cell-based in vitro assays. A Caenorhabditis elegans leaky gut model was established to investigate cream effects on gut health. Distinct differences were observed in peptide diversity and sequences released from HC-IMF compared with LC-IMF during simulated digestion (p < 0.05). Higher levels of free fatty acids were absorbed through 21-day differentiated Caco-2/HT-29MTX monolayers from HC-IMF, compared with LC-IMF and HBM (p < 0.05). Furthermore, the immune-modulating properties of HC-IMF appeared to be more similar to HBM than LC-IMF, as observed by comparable secretion of cytokines IL-10 and IL-1ß from THP-1 macrophages (p > 0.05). HC-IMF also supported intestinal recovery in C. elegans following distortion versus LC-IMF (p < 0.05). These observations suggest that cream as a lipid source in IMF may provide added nutritional and functional benefits more aligned with HBM.


Subject(s)
Caenorhabditis elegans , Digestion , Infant Formula , Milk, Human , Humans , Infant Formula/chemistry , Milk, Human/chemistry , Animals , Infant , Caco-2 Cells , Infant Nutritional Physiological Phenomena , HT29 Cells , Dairy Products , Intestines/physiology
17.
Nutrients ; 16(18)2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39339737

ABSTRACT

Mycotoxin contamination of cereals and cereal-based products is a serious problem for food safety. Antioxidant-rich ingredients such as bilberries (Vaccinium myrtillus L., VM) may mitigate their harmful effects. Firstly, total phenolic content, antioxidant activity, and analytical phytochemical composition (hydroxycinnamic and hydroxybenzoic acids, flavanols, flavonols, and anthocyanins) were assessed in lyophilized wild bilberries from Romania. Secondly, this study evaluated bilberries' effects on reducing ochratoxin A (OTA) bioaccessibility and cytotoxicity. An in vitro digestion model was developed and applied to four different types of bread: Control, VM (2%), OTA (15.89 ± 0.13 mg/kg), and OTA (16.79 ± 0.55 mg/kg)-VM (2%). The results indicated that VM decreased OTA bioaccessibility by 15% at the intestinal level. OTA-VM digests showed improved Caco-2 cell viability in comparison to OTA digests across different exposure times. Regarding the alterations in Jurkat cell line cell cycle phases and apoptosis/necrosis, significant increases in cell death were observed using OTA digests (11%), while VM addition demonstrated a protective effect (1%). Reactive oxygen species (ROS) analysis confirmed these findings, with OTA-VM digests showing significantly lower ROS levels compared to OTA digests, resulting in a 3.7-fold decrease. Thus, bilberries exhibit high potential as a functional ingredient, demonstrating protection in OTA mitigation effects.


Subject(s)
Antioxidants , Edible Grain , Ochratoxins , Ochratoxins/toxicity , Humans , Caco-2 Cells , Edible Grain/chemistry , Antioxidants/pharmacology , Vaccinium myrtillus/chemistry , Phytochemicals/pharmacology , Phytochemicals/analysis , Jurkat Cells , Cell Survival/drug effects , Reactive Oxygen Species/metabolism , Food Contamination/prevention & control , Food Contamination/analysis , Bread/analysis , Plant Extracts/pharmacology , Apoptosis/drug effects , Digestion/drug effects
18.
ACS Nano ; 18(39): 27026-27041, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39297569

ABSTRACT

Overcoming intestinal epithelial barriers to enhance bioavailability is a major challenge for oral delivery systems. Desirable nanocarriers should simultaneously exhibit rapid mucus penetration and efficient epithelial uptake; however, they two generally require contradictory structural properties. Herein, we proposed a strategy to construct multiperformance nanoparticles by modifying the rigidity of amphiphilic nanostructures originating from soy polypeptides (SPNPs), where its ability to overcome multibarriers was examined from both in vitro and in vivo, using curcumin (CUR) as a model cargo. Low-rigidity SPNPs showed higher affinity to mucin and were prone to getting stuck in the mucus layer. When they reached epithelial cells, they tended to be endocytosed through the clathrin and macropinocytosis pathways and further transferred to lysosomes, showing severe degradation and lower transport of CUR. Increased particle rigidity generally improved the absorption of CUR, with medium-rigidity SPNPs bloomed maximum plasma concentration of CUR by 80.62-fold and showed the highest oral bioavailability. Results from monocultured and cocultured cell models demonstrated that medium-rigidity SPNPs were least influenced by the mucus layer and changes in rigidity significantly influenced the endocytosis and intracellular fate of SPNPs. Those with higher rigidity preferred to be endocytosed via a caveolae-mediated pathway and trafficked to the ER and Golgi, facilitating their whole transcytosis, and avoiding intracellular metabolism. Moreover, rigidity modulation efficiently induces the reversible opening of intercellular tight junctions, which synergistically improves the transport of CUR into blood circulation. This study suggested that rigidity regulation on food originated amphiphilic peptides could overcome multiple physiological barriers, showing great potential as natural building block toward oral delivery.


Subject(s)
Biological Availability , Curcumin , Mucus , Curcumin/chemistry , Curcumin/pharmacokinetics , Curcumin/pharmacology , Curcumin/metabolism , Animals , Humans , Mucus/metabolism , Mucus/chemistry , Nanoparticles/chemistry , Nanoparticles/metabolism , Drug Carriers/chemistry , Caco-2 Cells , Particle Size , Endocytosis , Rats, Sprague-Dawley , Administration, Oral , Peptides/chemistry , Peptides/metabolism , Mice , Male
19.
Int J Mol Sci ; 25(18)2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39337545

ABSTRACT

Within class II bacteriocins, we assume the presence of a separate subfamily of antimicrobial peptides possessing a broad spectrum of antimicrobial activity. Although these peptides are structurally related to the subclass IIa (pediocin-like) bacteriocins, they have significant differences in biological activities and, probably, a mechanism of their antimicrobial action. A representative of this subfamily is acidocin A from Lactobacillus acidophilus TK9201. We discovered the similarity between acidocin A and acidocin 8912 from Lactobacillus acidophilus TK8912 when analyzing plasmids from lactic acid bacteria and suggested the presence of a single evolutionary predecessor of these peptides. We obtained the C-terminally extended homolog of acidocin 8912, named acidocin 8912A, a possible intermediate form in the evolution of the former. The study of secondary structures and biological activities of these peptides showed their structural similarity to acidocin A; however, the antimicrobial activities of acidocin 8912 and acidocin 8912A were lower than that of acidocin A. In addition, these peptides demonstrated stronger cytotoxic and membranotropic effects. Building upon what we previously discovered about the immunomodulatory properties of acidocin A, we studied its proteolytic stability under conditions simulating those in the digestive tract and also assessed its ability to permeate intestinal epithelium using the Caco-2 cells monolayer model. In addition, we found a pronounced effect of acidocin A against fungi of the genus Candida, which might also expand the therapeutic potential of this bacterial antimicrobial peptide.


Subject(s)
Bacteriocins , Lactobacillus acidophilus , Bacteriocins/chemistry , Bacteriocins/pharmacology , Bacteriocins/genetics , Humans , Lactobacillus acidophilus/drug effects , Amino Acid Sequence , Caco-2 Cells , Microbial Sensitivity Tests , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Hemolysis/drug effects , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/pharmacology , Protein Structure, Secondary , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry
20.
J Agric Food Chem ; 72(38): 21030-21040, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39283309

ABSTRACT

Mammalian milk exosomal miRNAs play an important role in maintaining intestinal immune homeostasis and protecting epithelial barrier function, but the specific miRNAs and whether miRNA-mediated mechanisms are responsible for these benefits remain a matter of investigation. This study isolated sheep milk-derived exosomes (sheep MDEs), identifying the enriched miRNAs in sheep MDEs, oar-miR-148a, and oar-let-7b as key components targeting TLR4 and TRAF1, which was validated by a dual-luciferase reporter assay. In dextran sulfate sodium-induced colitis mice, administration of sheep MDEs alleviated colitis symptoms, reduced colonic inflammation, and systemic oxidative stress, as well as significantly increased colonic oar-miR-148a and oar-let-7b while reducing toll-like receptor 4 (TLR4) and TNF-receptor-associated factor 1 (TRAF1) level. Further characterization in TNF-α-challenged Caco-2 cells showed that overexpression of these miRNAs suppressed the TLR4/TRAF1-IκBα-p65 pathway and reduced IL-6 and IL-12 production. These findings indicate that sheep MDEs exert gastrointestinal anti-inflammatory effects through the miRNA-mediated modulation of TLR4 and TRAF1, highlighting their potential in managing colitis.


Subject(s)
Colitis , Dextran Sulfate , Exosomes , MicroRNAs , Milk , TNF Receptor-Associated Factor 1 , Toll-Like Receptor 4 , Animals , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , MicroRNAs/immunology , Dextran Sulfate/adverse effects , Milk/chemistry , Milk/metabolism , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Mice , Sheep , Humans , Exosomes/genetics , Exosomes/metabolism , Exosomes/chemistry , Exosomes/immunology , TNF Receptor-Associated Factor 1/genetics , TNF Receptor-Associated Factor 1/metabolism , Caco-2 Cells , Male , Mice, Inbred C57BL , Female
SELECTION OF CITATIONS
SEARCH DETAIL