Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (31)

Search Parameters:
Keywords = Gut-Ex-Vivo System

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 17036 KiB  
Article
The Uremic Toxins Inorganic Phosphate, Indoxylsulphate, p-Cresylsulphate, and TMAO Induce the Generation of Sulphated Glycosaminoglycans in Aortic Tissue and Vascular Cells via pAKT Signaling: A Missing Link in the “Gut–Matrix Axis”
by Christian Freise, Susanne Metzkow, Andreas Zappe, Monika Ebert, Nicola Stolzenburg, Julia Hahndorf, Jörg Schnorr, Kevin Pagel and Matthias Taupitz
Toxins 2025, 17(5), 217; https://doi.org/10.3390/toxins17050217 - 25 Apr 2025
Viewed by 731
Abstract
Gut-derived uremic toxins (UTs) contribute to cardiovascular disorders like atherosclerosis and cardiomyopathy in patients with chronic kidney disease (CKD), causing increased cardiovascular morbidity and mortality. The intermediate steps between higher concentrations of gut-derived UTs and organ damage caused by UTs are still insufficiently [...] Read more.
Gut-derived uremic toxins (UTs) contribute to cardiovascular disorders like atherosclerosis and cardiomyopathy in patients with chronic kidney disease (CKD), causing increased cardiovascular morbidity and mortality. The intermediate steps between higher concentrations of gut-derived UTs and organ damage caused by UTs are still insufficiently understood. Glycosaminoglycans (GAGs) as components of the extracellular matrix are known to interact with various ligands such as growth factors or receptors, thereby influencing (patho)physiological processes. We previously found that the UT inorganic phosphate (Pi) induces the synthesis and sulphation of the GAGs heparan sulphate and chondroitin sulphate in the rat vascular smooth muscle cell (VSMC) line A7r5 and in the human endothelial cell (EC) line EA.Hy926. The aim of this study was to investigate if other organic UTs modulate GAGs in vascular cells as well. We treated ex vivo cultures of rat aortic rings as well as primary rat VSMCs and human ECs with the UTs Pi, indoxylsulphate (IS), p-cresylsulphate (pCS), trimethylamine N-oxide (TMAO), and urea, and analyzed the samples by histological staining, qPCR, western blot, HPLC, and colorimetric assays. The UT treatment of aortic rings and cells increased contents of sulphated GAGs and hyaluronic acid. UT-treated cells contained higher amounts of 4S- and 6S-sulphated GAGs compared to controls. This was accompanied by altered expressions of genes and proteins relevant for GAG metabolism. Mechanistically, the effects of the UTs on GAGs involve the activation of the PI3K/Akt pathway and of the transcription factor NF-κB. In conclusion, the UT-induced remodeling of the cardiovascular matrix by upregulation of sulphated GAGs and hyaluronic acid in aortic tissue and vascular cells might be a missing link between gut-derived UT and pathophysiological alterations in the cardiovascular system in the sense of a gut–matrix axis. Full article
Show Figures

Figure 1

23 pages, 2265 KiB  
Review
The Western Diet and Atopic Dermatitis: The Potential Role of Nutrients, Contaminants, and Additives in Dysbiosis and Epithelial Barrier Dysfunction
by Chiara Maria Teresa Boggio, Federica Veronese, Marta Armari, Elisa Zavattaro, Elia Esposto, Paola Savoia and Barbara Azzimonti
Antioxidants 2025, 14(4), 386; https://doi.org/10.3390/antiox14040386 - 25 Mar 2025
Cited by 1 | Viewed by 2776
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disorder influenced by both genetic and environmental factors, collectively termed the exposome. Among these determinants, diet emerges as a pivotal component, with diverse nutrients, contaminants, and additives shaping immune responses, microbiota composition, and systemic inflammatory [...] Read more.
Atopic dermatitis (AD) is a chronic inflammatory skin disorder influenced by both genetic and environmental factors, collectively termed the exposome. Among these determinants, diet emerges as a pivotal component, with diverse nutrients, contaminants, and additives shaping immune responses, microbiota composition, and systemic inflammatory status. This literature review aimed to elucidate the interplay between dietary factors and skin dysbiosis in AD, providing insights into how these interactions may impact disease susceptibility and progression. A comprehensive search of PubMed and Scopus was conducted using relevant keywords and medical subject headings (MeSH). Studies published in English within the past 25 years were included, encompassing in vitro, in vivo, and ex vivo research, as well as reviews. Priority was given to frequently cited articles, reflecting significant contributions to current understanding. Findings suggest that dietary habits influence AD by modulating both gut and skin microbiota, immune pathways, and inflammatory processes. These insights underscore the importance of considering diet within a broader exposome framework, paving the way for targeted interventions to improve AD management. Further research is needed to clarify the mechanisms and optimize nutritional strategies, potentially informing preventive and therapeutic approaches for AD. Full article
(This article belongs to the Special Issue Interplay of Microbiome and Oxidative Stress)
Show Figures

Figure 1

13 pages, 2168 KiB  
Article
Carrot-Derived Rhamnogalacturonan-I Consistently Increases the Microbial Production of Health-Promoting Indole-3-Propionic Acid Ex Vivo
by Annick Mercenier, Lam Dai Vu, Jonas Poppe, Ruud Albers, Sue McKay and Pieter Van den Abbeele
Metabolites 2024, 14(12), 722; https://doi.org/10.3390/metabo14120722 - 21 Dec 2024
Viewed by 2114
Abstract
Background: Using dietary interventions to steer the metabolic output of the gut microbiota towards specific health-promoting metabolites is often challenging due to interpersonal variation in treatment responses. Methods: In this study, we combined the ex vivo SIFR® (Systemic Intestinal Fermentation Research) technology [...] Read more.
Background: Using dietary interventions to steer the metabolic output of the gut microbiota towards specific health-promoting metabolites is often challenging due to interpersonal variation in treatment responses. Methods: In this study, we combined the ex vivo SIFR® (Systemic Intestinal Fermentation Research) technology with untargeted metabolite profiling to investigate the impact of carrot-derived rhamnogalacturonan-I (cRG-I) on ex vivo metabolite production by the gut microbiota of 24 human adults. Results: The findings reveal that at a dose equivalent to 1.5 g/d, cRG-I consistently promoted indole-3-propionic acid (IPA) production (+45.8% increase) across all subjects. At a dose equivalent to 0.3 g/d, increased IPA production was also observed (+14.6%), which was comparable to the effect seen for 1.5 g/d inulin (10.6%). IPA has been shown to provide protection against diseases affecting the gut and multiple organs. The Pearson correlation analysis revealed a strong correlation (R = 0.65, padjusted = 6.1 × 10−16) between the increases in IPA levels and the absolute levels of Bifidobacterium longum, a producer of indole-3-lactic acid (ILA), an intermediate in IPA production. Finally, the community modulation score, a novel diversity index, demonstrated that cRG-I maintained a high α-diversity which has previously been linked to elevated IPA production. Conclusions: The results from the ex vivo SIFR® experiment mirrored clinical outcomes and provided novel insights into the impact of cRG-I on the gut microbiome function. Importantly, we demonstrated that cRG-I promotes tryptophan conversion into IPA via gut microbiome modulation, thus conferring benefits via amino acid derived metabolites extending beyond those previously reported for short chain fatty acids (SCFA) resulting from carbohydrate fermentation. Full article
Show Figures

Graphical abstract

16 pages, 2939 KiB  
Article
In Vitro Mucoadhesive Features of Gliadin Nanoparticles Containing Thiamine Hydrochloride
by Silvia Voci, Agnese Gagliardi, Elena Giuliano, Maria Cristina Salvatici, Antonio Procopio and Donato Cosco
Pharmaceutics 2024, 16(10), 1296; https://doi.org/10.3390/pharmaceutics16101296 - 4 Oct 2024
Cited by 1 | Viewed by 1374
Abstract
Background: Gliadins have aroused significant interest in the last decade as suitable biomaterials for food and pharmaceutical applications. In particular, the oral route is the preferred method of administration for gliadin-based formulations, due to the affinity of this biomaterial for the gut mucosa. [...] Read more.
Background: Gliadins have aroused significant interest in the last decade as suitable biomaterials for food and pharmaceutical applications. In particular, the oral route is the preferred method of administration for gliadin-based formulations, due to the affinity of this biomaterial for the gut mucosa. However, up to now, this has been demonstrated only by means of in vivo or ex vivo studies. Methods: This is why, in this study, various in vitro techniques were employed in order to evaluate the ability of polymeric nanoparticles, made up of a commercial grade of the protein and an etheric surfactant, to interact with porcine gastric mucin. The nanosystems were also used for the encapsulation of thiamine hydrochloride, used as a model of a micronutrient. Results: The resulting systems were characterized by a mean diameter of ~160–170 nm, a narrow size distribution when 0.2–0.6 mg/mL of thiamine was used, and an encapsulation efficiency between 30 and 45% of the drug initially employed. The incubation of the gliadin nanosystems with various concentrations of porcine gastric mucin evidenced the ability of the carriers to interact with the mucus glycoprotein, showing a decreased Zeta potential after a 4 h incubation (from ~−30 to −40 mV), while demonstrating that the encapsulation of the drug did not affect its bioadhesive features. Conclusions: Altogether, these data support the conceivable application of gliadin nanoparticles as formulations for the oral administration of bioactive compounds. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Graphical abstract

20 pages, 4220 KiB  
Review
Upgrading In Vitro Digestion Protocols with Absorption Models
by Otilia Antal, István Dalmadi and Krisztina Takács
Appl. Sci. 2024, 14(18), 8320; https://doi.org/10.3390/app14188320 - 15 Sep 2024
Cited by 3 | Viewed by 2144
Abstract
Intestinal digestion and absorption are complex processes; thus, it is a challenge to imitate them realistically. There are numerous approaches available, with different disadvantages and advantages. The simplest methods to mimic absorption are the non-cell-based transport models but these lack important characteristics of [...] Read more.
Intestinal digestion and absorption are complex processes; thus, it is a challenge to imitate them realistically. There are numerous approaches available, with different disadvantages and advantages. The simplest methods to mimic absorption are the non-cell-based transport models but these lack important characteristics of enterocytes of the intestine. Therefore, the most often used method is to measure absorption through viable mammalian cells (most commonly Caco-2 cells, cultured on membrane insert plates), which not only assures the incorporation of brush border enzymes (responsible for the final digestion of peptides and disaccharides), it also simulates the absorption process. This means that influx/efflux transporter-facilitated transport, carrier-mediated transport, endocytosis, and transcytosis is also imitated besides passive diffusion. Still, these also lack the complexity of intestinal epithelium. Organoids or ex vivo models are a better approach if we want to attain precision but the highest accuracy can be achieved with microfluidic systems (gut-on-a-chip models). We propose that more research is necessary, and food absorption should also be studied on gut-on-a-chips, especially with fragmented organoids. Our review supports the choices of a proper intestinal epithelium model, which may have a key role in functional food development, nutrition studies, and toxicity assessment. Full article
(This article belongs to the Special Issue Feature Review Papers in Section ‘Food Science and Technology')
Show Figures

Figure 1

33 pages, 10873 KiB  
Review
Effects of Cannabinoids on Intestinal Motility, Barrier Permeability, and Therapeutic Potential in Gastrointestinal Diseases
by Kijan Crowley, Łukasz Kiraga, Edyta Miszczuk, Sergiusz Skiba, Joanna Banach, Urszula Latek, Marta Mendel and Magdalena Chłopecka
Int. J. Mol. Sci. 2024, 25(12), 6682; https://doi.org/10.3390/ijms25126682 - 18 Jun 2024
Cited by 12 | Viewed by 6263
Abstract
Cannabinoids and their receptors play a significant role in the regulation of gastrointestinal (GIT) peristalsis and intestinal barrier permeability. This review critically evaluates current knowledge about the mechanisms of action and biological effects of endocannabinoids and phytocannabinoids on GIT functions and the potential [...] Read more.
Cannabinoids and their receptors play a significant role in the regulation of gastrointestinal (GIT) peristalsis and intestinal barrier permeability. This review critically evaluates current knowledge about the mechanisms of action and biological effects of endocannabinoids and phytocannabinoids on GIT functions and the potential therapeutic applications of these compounds. The results of ex vivo and in vivo preclinical data indicate that cannabinoids can both inhibit and stimulate gut peristalsis, depending on various factors. Endocannabinoids affect peristalsis in a cannabinoid (CB) receptor-specific manner; however, there is also an important interaction between them and the transient receptor potential cation channel subfamily V member 1 (TRPV1) system. Phytocannabinoids such as Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD) impact gut motility mainly through the CB1 receptor. They were also found to improve intestinal barrier integrity, mainly through CB1 receptor stimulation but also via protein kinase A (PKA), mitogen-associated protein kinase (MAPK), and adenylyl cyclase signaling pathways, as well as by influencing the expression of tight junction (TJ) proteins. The anti-inflammatory effects of cannabinoids in GIT disorders are postulated to occur by the lowering of inflammatory factors such as myeloperoxidase (MPO) activity and regulation of cytokine levels. In conclusion, there is a prospect of utilizing cannabinoids as components of therapy for GIT disorders. Full article
(This article belongs to the Special Issue Natural-Derived Bioactive Compounds in Disease Treatment)
Show Figures

Figure 1

17 pages, 4567 KiB  
Article
Intestinal Ketogenesis and Permeability
by Anna Casselbrant, Erik Elias, Peter Hallersund, Erik Elebring, Jakob Cervin, Lars Fändriks and Ville Wallenius
Int. J. Mol. Sci. 2024, 25(12), 6555; https://doi.org/10.3390/ijms25126555 - 14 Jun 2024
Cited by 1 | Viewed by 2324
Abstract
Consumption of a high-fat diet (HFD) has been suggested as a contributing factor behind increased intestinal permeability in obesity, leading to increased plasma levels of microbial endotoxins and, thereby, increased systemic inflammation. We and others have shown that HFD can induce jejunal expression [...] Read more.
Consumption of a high-fat diet (HFD) has been suggested as a contributing factor behind increased intestinal permeability in obesity, leading to increased plasma levels of microbial endotoxins and, thereby, increased systemic inflammation. We and others have shown that HFD can induce jejunal expression of the ketogenic rate-limiting enzyme mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase (HMGCS). HMGCS is activated via the free fatty acid binding nuclear receptor PPAR-α, and it is a key enzyme in ketone body synthesis that was earlier believed to be expressed exclusively in the liver. The function of intestinal ketogenesis is unknown but has been described in suckling rats and mice pups, possibly in order to allow large molecules, such as immunoglobulins, to pass over the intestinal barrier. Therefore, we hypothesized that ketone bodies could regulate intestinal barrier function, e.g., via regulation of tight junction proteins. The primary aim was to compare the effects of HFD that can induce intestinal ketogenesis to an equicaloric carbohydrate diet on inflammatory responses, nutrition sensing, and intestinal permeability in human jejunal mucosa. Fifteen healthy volunteers receiving a 2-week HFD diet compared to a high-carbohydrate diet were compared. Blood samples and mixed meal tests were performed at the end of each dietary period to examine inflammation markers and postprandial endotoxemia. Jejunal biopsies were assessed for protein expression using Western blotting, immunohistochemistry, and morphometric characteristics of tight junctions by electron microscopy. Functional analyses of permeability and ketogenesis were performed in Caco-2 cells, mice, and human enteroids. Ussing chambers were used to analyze permeability. CRP and ALP values were within normal ranges and postprandial endotoxemia levels were low and did not differ between the two diets. The PPARα receptor was ketone body-dependently reduced after HFD. None of the tight junction proteins studied, nor the basal electrical parameters, were different between the two diets. However, the ketone body inhibitor hymeglusin increased resistance in mucosal biopsies. In addition, the tight junction protein claudin-3 was increased by ketone inhibition in human enteroids. The ketone body β-Hydroxybutyrate (βHB) did not, however, change the mucosal transition of the large-size molecular FD4-probe or LPS in Caco-2 and mouse experiments. We found that PPARα expression was inhibited by the ketone body βHB. As PPARα regulates HMGCS expression, the ketone bodies thus exert negative feedback signaling on their own production. Furthermore, ketone bodies were involved in the regulation of permeability on intestinal mucosal cells in vitro and ex vivo. We were not, however, able to reproduce these effects on intestinal permeability in vivo in humans when comparing two weeks of high-fat with high-carbohydrate diet in healthy volunteers. Further, neither the expression of inflammation markers nor the aggregate tight junction proteins were changed. Thus, it seems that not only HFD but also other factors are needed to permit increased intestinal permeability in vivo. This indicates that the healthy gut can adapt to extremes of macro-nutrients and increased levels of intestinally produced ketone bodies, at least during a shorter dietary challenge. Full article
(This article belongs to the Special Issue The Role of Tight Junction Proteins in Health and Disease)
Show Figures

Figure 1

22 pages, 4193 KiB  
Article
The Effect of Opuntia ficus Mucilage Pectin and Citrus aurantium Extract Added to a Food Matrix on the Gut Microbiota of Lean Humans and Humans with Obesity
by Nancy Abril Estrada-Sierra, Marisela Gonzalez-Avila, Judith-Esmeralda Urias-Silvas, Gabriel Rincon-Enriquez, Maria Dolores Garcia-Parra and Socorro Josefina Villanueva-Rodriguez
Foods 2024, 13(4), 587; https://doi.org/10.3390/foods13040587 - 15 Feb 2024
Cited by 1 | Viewed by 2091
Abstract
Experimental studies have provided evidence that physicochemical interactions in the food matrix can modify the biologically beneficial effects of bioactive compounds, including their effect on gut microbiota. This work aimed to evaluate the effect of a food gel matrix with Opuntia ficus cladodes [...] Read more.
Experimental studies have provided evidence that physicochemical interactions in the food matrix can modify the biologically beneficial effects of bioactive compounds, including their effect on gut microbiota. This work aimed to evaluate the effect of a food gel matrix with Opuntia ficus cladodes mucilage pectin and Citrus Aurantium extract on the growth of four beneficial gut bacteria obtained from the fecal microbiota of people who are lean or who have obesity after digestion in the upper digestive system. To accomplish this, a base formulation of Opuntia ficus cladodes mucilage with or without C. aurantium extract was submitted to an ex vivo fecal fermentation in an automatic and robotic intestinal system. The changes in the intestinal microbiota were determined by means of plate culture and 16S sequencing, while short-chain fatty acids (SCFA) produced in the colon were determined via gas chromatography. In the presence of the extract in formulation, greater growth of Bifidobacterium spp. (+1.6 Log10 Colonic Forming Unit, UFC) and Lactobacillus spp. (+2 Log10 UFC) in the microbiota of lean people was observed. Only the growth in Salmonella spp. (−1 Log10 UFC) from both microbiota was affected in the presence of the extract, which decreased in the ascending colon. SCFA was mainly produced by the microbiota of people who were lean rather than those who had obesity in the presence of the extract, particularly in the ascending colon. The effect of sour orange extract seems to depend on the origin of the microbiota, whether in people who have obesity (25 mM/L) or are lean (39 mM/L). Full article
(This article belongs to the Section Nutraceuticals, Functional Foods, and Novel Foods)
Show Figures

Graphical abstract

13 pages, 1020 KiB  
Article
Deoxycholic Acid, a Secondary Bile Acid, Increases Cardiac Output and Blood Pressure in Rats
by Artur Nowiński, Dawid Chabowski, Joanna Giebułtowicz, Marta Aleksandrowicz and Marcin Ufnal
Nutrients 2024, 16(1), 32; https://doi.org/10.3390/nu16010032 - 21 Dec 2023
Cited by 7 | Viewed by 2452
Abstract
Background: Deoxycholic acid (DCA) is a secondary bile acid produced by gut bacteria. Elevated serum concentrations of DCA are observed in cardiovascular disease (CVD). We hypothesized that DCA might influence hemodynamic parameters in rats. Methods: The concentration of DCA in systemic blood was [...] Read more.
Background: Deoxycholic acid (DCA) is a secondary bile acid produced by gut bacteria. Elevated serum concentrations of DCA are observed in cardiovascular disease (CVD). We hypothesized that DCA might influence hemodynamic parameters in rats. Methods: The concentration of DCA in systemic blood was measured with liquid chromatography coupled with mass spectrometry. Arterial blood pressure (BP), heart rate (HR) and echocardiographic parameters were evaluated in anesthetized, male, 3–4-month-old Sprague–Dawley rats administered intravenously (IV) or intracerebroventricularly (ICV) with investigated compounds. Mesenteric artery (MA) reactivity was tested ex vivo. Results: The baseline plasma concentration of DCA was 0.24 ± 0.03 mg/L. The oral antibiotic treatment produced a large decrease in the concentration. Administered IV, the compound increased BP and HR in a dose-dependent manner. DCA also increased heart contractility and cardiac output. None of the tested compounds—prazosin (an alpha-blocker), propranolol (beta-adrenolytic), atropine (muscarinic receptor antagonist), glibenclamide (K-ATP inhibitor) or DY 268 (FXR antagonist), glycyrrhetinic acid (11HSD2 inhibitor)—significantly diminished the DCA-induced pressor effect. ICV infusion did not exert significant HR or BP changes. DCA relaxed MAs. Systemic vascular resistance did not change significantly. Conclusions: DCA elevates BP primarily by augmenting cardiac output. As a metabolite derived from gut bacteria, DCA potentially serves as a mediator in the interaction between the gut microbiota and the host’s circulatory system. Full article
(This article belongs to the Special Issue Nutritional Status and Cardiovascular Diseases)
Show Figures

Figure 1

13 pages, 3330 KiB  
Article
Prevention of Colitis-Associated Cancer via Oral Administration of M13-Loaded Lipid Nanoparticles
by Dingpei Long, Zahra Alghoul, Junsik Sung, Chunhua Yang and Didier Merlin
Pharmaceutics 2023, 15(9), 2331; https://doi.org/10.3390/pharmaceutics15092331 - 16 Sep 2023
Cited by 3 | Viewed by 6623
Abstract
Inflammatory bowel disease (IBD), which includes ulcerative colitis (UC) and Crohn’s disease, is known to increase the risk of colitis-associated cancer (CAC). CAC has been found to be unresponsive to standard chemotherapy regimens, and the current treatments do not utilize effective small-molecule drugs [...] Read more.
Inflammatory bowel disease (IBD), which includes ulcerative colitis (UC) and Crohn’s disease, is known to increase the risk of colitis-associated cancer (CAC). CAC has been found to be unresponsive to standard chemotherapy regimens, and the current treatments do not utilize effective small-molecule drugs and colon-targeted delivery systems. Previous studies indicated that the M13–nano-liposome (NL) formulation can effectively target the colon and reshape the gut microbiota in ex vivo cultures, generating altered microbial metabolites that can efficiently prevent chronic UC. In this study, we tested the cancer cell uptake ability of the NL formulation and investigated the potential of the M13–NL formulation to prevent CAC in the azoxymethane (AOM)-exposed IL10−/− mouse model. Our findings demonstrate that oral administration of M13–NL prevents tumor development in AOM-exposed IL10−/− mice, suggesting that M13–NL is a promising oral drug formulation for preventing CAC. Full article
Show Figures

Graphical abstract

27 pages, 493 KiB  
Review
Intestinal Barrier Dysfunction and Microbiota–Gut–Brain Axis: Possible Implications in the Pathogenesis and Treatment of Autism Spectrum Disorder
by Vanessa Nadia Dargenio, Costantino Dargenio, Stefania Castellaneta, Andrea De Giacomo, Marianna Laguardia, Federico Schettini, Ruggiero Francavilla and Fernanda Cristofori
Nutrients 2023, 15(7), 1620; https://doi.org/10.3390/nu15071620 - 27 Mar 2023
Cited by 50 | Viewed by 8808
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder with multifactorial etiology, characterized by impairment in two main functional areas: (1) communication and social interactions, and (2) skills, interests and activities. ASD patients often suffer from gastrointestinal symptoms associated with dysbiotic states and [...] Read more.
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder with multifactorial etiology, characterized by impairment in two main functional areas: (1) communication and social interactions, and (2) skills, interests and activities. ASD patients often suffer from gastrointestinal symptoms associated with dysbiotic states and a “leaky gut.” A key role in the pathogenesis of ASD has been attributed to the gut microbiota, as it influences central nervous system development and neuropsychological and gastrointestinal homeostasis through the microbiota–gut–brain axis. A state of dysbiosis with a reduction in the Bacteroidetes/Firmicutes ratio and Bacteroidetes level and other imbalances is common in ASD. In recent decades, many authors have tried to study and identify the microbial signature of ASD through in vivo and ex vivo studies. In this regard, the advent of metabolomics has also been of great help. Based on these data, several therapeutic strategies, primarily the use of probiotics, are investigated to improve the symptoms of ASD through the modulation of the microbiota. However, although the results are promising, the heterogeneity of the studies precludes concrete evidence. The aim of this review is to explore the role of intestinal barrier dysfunction, the gut–brain axis and microbiota alterations in ASD and the possible role of probiotic supplementation in these patients. Full article
15 pages, 3379 KiB  
Article
Lacticaseibacillus rhamnosus ATCC 53103 and Limosilactobacillus reuteri ATCC 53608 Synergistically Boost Butyrate Levels upon Tributyrin Administration Ex Vivo
by Pieter Van den Abbeele, Mallory Goggans, Stef Deyaert, Aurélien Baudot, Michiel Van de Vliet, Marta Calatayud Arroyo and Michael Lelah
Int. J. Mol. Sci. 2023, 24(6), 5859; https://doi.org/10.3390/ijms24065859 - 20 Mar 2023
Cited by 7 | Viewed by 3667
Abstract
Modulation of the gut microbiota is a trending strategy to improve health. While butyrate has been identified as a key health-related microbial metabolite, managing its supply to the host remains challenging. Therefore, this study investigated the potential to manage butyrate supply via tributyrin [...] Read more.
Modulation of the gut microbiota is a trending strategy to improve health. While butyrate has been identified as a key health-related microbial metabolite, managing its supply to the host remains challenging. Therefore, this study investigated the potential to manage butyrate supply via tributyrin oil supplementation (TB; glycerol with three butyrate molecules) using the ex vivo SIFR® (Systemic Intestinal Fermentation Research) technology, a highly reproducible, in vivo predictive gut model that accurately preserves in vivo-derived microbiota and enables addressing interpersonal differences. Dosing 1 g TB/L significantly increased butyrate with 4.1 (±0.3) mM, corresponding with 83 ± 6% of the theoretical butyrate content of TB. Interestingly, co-administration of Limosilactobacillus reuteri ATCC 53608 (REU) and Lacticaseibacillus rhamnosus ATCC 53103 (LGG) markedly enhanced butyrate to levels that exceeded the theoretical butyrate content of TB (138 ± 11% for REU; 126 ± 8% for LGG). Both TB + REU and TB + LGG stimulated Coprococcus catus, a lactate-utilizing, butyrate-producing species. The stimulation of C. catus with TB + REU was remarkably consistent across the six human adults tested. It is hypothesized that LGG and REU ferment the glycerol backbone of TB to produce lactate, a precursor of butyrate. TB + REU also significantly stimulated the butyrate-producing Eubacterium rectale and Gemmiger formicilis and promoted microbial diversity. The more potent effects of REU could be due to its ability to convert glycerol to reuterin, an antimicrobial compound. Overall, both the direct butyrate release from TB and the additional butyrate production via REU/LGG-mediated cross-feeding were highly consistent. This contrasts with the large interpersonal differences in butyrate production that are often observed upon prebiotic treatment. Combining TB with LGG and especially REU is thus a promising strategy to consistently supply butyrate to the host, potentially resulting in more predictable health benefits. Full article
(This article belongs to the Special Issue Gut Microbes and Their Metabolites in Health and Disease)
Show Figures

Graphical abstract

18 pages, 5061 KiB  
Article
Group I Metabotropic Glutamate Receptors Modulate Motility and Enteric Neural Activity in the Mouse Colon
by Anita J. L. Leembruggen, Yuqing Lu, Haozhe Wang, Volkan Uzungil, Thibault Renoir, Anthony J. Hannan, Lincon A. Stamp, Marlene M. Hao and Joel C. Bornstein
Biomolecules 2023, 13(1), 139; https://doi.org/10.3390/biom13010139 - 9 Jan 2023
Cited by 4 | Viewed by 2857
Abstract
Glutamate is the major excitatory neurotransmitter in the central nervous system, and there is evidence that Group-I metabotropic glutamate receptors (mGlu1 and mGlu5) have established roles in excitatory neurotransmission and synaptic plasticity. While glutamate is abundantly present in the gut, it plays a [...] Read more.
Glutamate is the major excitatory neurotransmitter in the central nervous system, and there is evidence that Group-I metabotropic glutamate receptors (mGlu1 and mGlu5) have established roles in excitatory neurotransmission and synaptic plasticity. While glutamate is abundantly present in the gut, it plays a smaller role in neurotransmission in the enteric nervous system. In this study, we examined the roles of Group-I mGlu receptors in gastrointestinal function. We investigated the expression of Grm1 (mGlu1) and Grm5 (mGlu5) in the mouse myenteric plexus using RNAscope in situ hybridization. Live calcium imaging and motility analysis were performed on ex vivo preparations of the mouse colon. mGlu5 was found to play a role in excitatory enteric neurotransmission, as electrically-evoked calcium transients were sensitive to the mGlu5 antagonist MPEP. However, inhibition of mGlu5 activity did not affect colonic motor complexes (CMCs). Instead, inhibition of mGlu1 using BAY 36-7620 reduced CMC frequency but did not affect enteric neurotransmission. These data highlight complex roles for Group-I mGlu receptors in myenteric neuron activity and colonic function. Full article
(This article belongs to the Special Issue Enteric Nervous System: Normal Functions and Enteric Neuropathies)
Show Figures

Figure 1

17 pages, 2985 KiB  
Article
Bioadhesive Tannic-Acid-Functionalized Zein Coating Achieves Engineered Colonic Delivery of IBD Therapeutics via Reservoir Microdevices
by Khorshid Kamguyan, Rolf Bech Kjeldsen, Saeed Zajforoushan Moghaddam, Melanie Randahl Nielsen, Esben Thormann, Kinga Zór, Line Hagner Nielsen and Anja Boisen
Pharmaceutics 2022, 14(11), 2536; https://doi.org/10.3390/pharmaceutics14112536 - 21 Nov 2022
Cited by 13 | Viewed by 3243
Abstract
The biggest challenge in oral delivery of anti-inflammatory drugs such as 5-aminosalicylic acid (5-ASA) is to (i) prevent rapid absorption in the small intestine and (ii) achieve localized release at the site of inflammation in the lower gut, i.e., the colon. Here, we [...] Read more.
The biggest challenge in oral delivery of anti-inflammatory drugs such as 5-aminosalicylic acid (5-ASA) is to (i) prevent rapid absorption in the small intestine and (ii) achieve localized release at the site of inflammation in the lower gut, i.e., the colon. Here, we present an advanced biopolymeric coating comprising of tannic-acid-functionalized zein protein to provide a sustained, colon-targeted release profile for 5-ASA and enhance the mucoadhesion of the dosage form via a mussel-inspired mechanism. To enable localized delivery and provide high local concentration, 5-ASA is loaded into the microfabricated drug carriers (microcontainers) and sealed with the developed coating. The functionality and drug release profile of the coating are characterized and optimized in vitro, showing great tunability, scalability, and stability toward proteases. Further, ex vivo experiments demonstrate that the tannic acid functionalization can significantly enhance the mucoadhesion of the coating, which is followed up by in vivo investigations on the intestinal retention, and pharmacokinetic evaluation of the 5-ASA delivery system. Results indicate that the developed coating can provide prolonged colonic delivery of 5-ASA. Therefore, the here-developed biodegradable coating can be an eco-friendly substitute to the state-of-the-art commercial counterparts for targeted delivery of 5-ASA and other small molecule drugs. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Graphical abstract

17 pages, 1983 KiB  
Communication
The Gut-Ex-Vivo System (GEVS) Is a Dynamic and Versatile Tool for the Study of DNBS-Induced IBD in BALB/C and C57BL/6 Mice, Highlighting the Protective Role of Probiotics
by Romina Monzani, Mara Gagliardi, Nausicaa Clemente, Valentina Saverio, Elżbieta Pańczyszyn, Claudio Santoro, Nissan Yissachar, Annalisa Visciglia, Marco Pane, Angela Amoruso and Marco Corazzari
Biology 2022, 11(11), 1574; https://doi.org/10.3390/biology11111574 - 27 Oct 2022
Cited by 5 | Viewed by 3235
Abstract
Background: IBD is a spectrum of pathologies characterized by dysregulated immune activation leading to uncontrolled response against the intestine, thus resulting in chronic gut inflammation and tissue damage. Due to its complexity, the molecular mechanisms responsible for disease onset and progression are still [...] Read more.
Background: IBD is a spectrum of pathologies characterized by dysregulated immune activation leading to uncontrolled response against the intestine, thus resulting in chronic gut inflammation and tissue damage. Due to its complexity, the molecular mechanisms responsible for disease onset and progression are still elusive, thus requiring intense research effort. In this context, the development of models replicating the etiopathology of IBD and allowing the testing of new potential therapies is critical. Methods: Colon from C57BL/6 or BALB/c mice was cultivated in a Gut-Ex-Vivo System (GEVS), exposed for 5 h to DNBS 1.5 or 2.5 mg/mL, in presence or absence of two probiotic formulations (P1 = Bifidobacterium breve BR03 (DSM16604) and B632 (DSM24706); P2 = Lacticaseibacillus rhamnosus LR04 (DSM16605), Lactiplantibacillus plantarum LP14 (DSM33401) and Lacticaseibacillus paracasei LPC09), and the main hallmarks of IBD were evaluated. Results: Gene expression analysis revealed the following DNBS-induced effects: (i) compromised tight junction organization, responsible for tissue permeability dysregulation; (ii) induction of ER stress, and (iii) tissue inflammation in colon of C57BL/6 mice. Moreover, the concomitant DNBS-induced apoptosis and ferroptosis pathways were evident in colon from both BALB/c and C57BL/6 mice. Finally, the co-administration of probiotics completely prevented the detrimental effects of DNBS. Conclusions: Overall, we have provided results demonstrating that GEVS is a consistent, reliable, and cost-effective system for modeling DNBS-induced IBD, useful for studying the onset and progression of human disease at the molecular level, while also reducing animal suffering. Moreover, we have confirmed the beneficial effect of probiotics administration in promoting the remission of IBD. Full article
Show Figures

Graphical abstract

Back to TopTop