Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.848
Filter
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167221, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718845

ABSTRACT

BACKGROUND: Short bowel syndrome (SBS) features nutrients malabsorption and impaired intestinal barrier. Patients with SBS are prone to sepsis, intestinal flora dysbiosis and intestinal failure associated liver disease. Protecting intestinal barrier and preventing complications are potential strategies for SBS treatment. This study aims to investigate the effects of farnesoid X receptor (FXR) agonist, obeticholic acid (OCA), have on intestinal barrier and ecological environment in SBS. METHODS AND RESULTS: Through testing the small intestine and serum samples of patients with SBS, impaired intestinal barrier was verified, as evidenced by reduced expressions of intestinal tight junction proteins (TJPs), increased levels of apoptosis and epithelial cell damage. The intestinal expressions of FXR and related downstream molecules were decreased in SBS patients. Then, global FXR activator OCA was used to further dissect the potential role of the FXR in a rat model of SBS. Low expressions of FXR-related molecules were observed on the small intestine of SBS rats, along with increased proinflammatory factors and damaged barrier function. Furthermore, SBS rats possessed significantly decreased body weight and elevated death rate. Supplementation with OCA mitigated the damaged intestinal barrier and increased proinflammatory factors in SBS rats, accompanied by activated FXR-related molecules. Using 16S rDNA sequencing, the regulatory role of OCA on gut microbiota in SBS rats was witnessed. LPS stimulation to Caco-2 cells induced apoptosis and overexpression of proinflammatory factors in vitro. OCA incubation of LPS-pretreated Caco-2 cells activated FXR-related molecules, increased the expressions of TJPs, ameliorated apoptosis and inhibited overexpression of proinflammatory factors. CONCLUSIONS: OCA supplementation could effectively ameliorate the intestinal barrier disruption and inhibit overexpression of proinflammatory factors in a rat model of SBS and LPS-pretreated Caco-2 cells. As a selective activator of FXR, OCA might realize its protective function through FXR activation.


Subject(s)
Chenodeoxycholic Acid , Disease Models, Animal , Intestinal Mucosa , Receptors, Cytoplasmic and Nuclear , Short Bowel Syndrome , Animals , Chenodeoxycholic Acid/analogs & derivatives , Chenodeoxycholic Acid/pharmacology , Short Bowel Syndrome/metabolism , Short Bowel Syndrome/drug therapy , Short Bowel Syndrome/pathology , Rats , Humans , Male , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Gastrointestinal Microbiome/drug effects , Female , Rats, Sprague-Dawley , Apoptosis/drug effects , Middle Aged , Intestine, Small/metabolism , Intestine, Small/drug effects , Intestine, Small/pathology , Adult , Tight Junction Proteins/metabolism
2.
Ecotoxicol Environ Saf ; 276: 116270, 2024 May.
Article in English | MEDLINE | ID: mdl-38574645

ABSTRACT

Mycotoxin contamination has become a major food safety issue and greatly threatens human and animal health. Patulin (PAT), a common mycotoxin in the environment, is exposed through the food chain and damages the gastrointestinal tract. However, its mechanism of enterotoxicity at the genetic and metabolic levels remains to be elucidated. Herein, the intestinal histopathological and biochemical indices, transcriptome, and metabolome of C57BL/6 J mice exposed to different doses of PAT were successively assessed, as well as the toxicokinetics of PAT in vivo. The results showed that acute PAT exposure induced damaged villi and crypts, reduced mucus secretion, decreased SOD and GSH-Px activities, and enhanced MPO activity in the small intestine and mild damage in the colon. At the transcriptional level, the genes affected by PAT were dose-dependently altered in the small intestine and fluctuated in the colon. PAT primarily affected inflammation-related signaling pathways and oxidative phosphorylation in the small intestine and immune responses in the colon. At the metabolic level, amino acids decreased, and extensive lipids accumulated in the small intestine and colon. Seven metabolic pathways were jointly affected by PAT in two intestinal sites. Moreover, changes in PAT products and GST activity were detected in the small intestinal tissue but not in the colonic tissue, explaining the different damage degrees of the two sites. Finally, the integrated results collectively explained the toxicological mechanism of PAT, which damaged the small intestine directly and the colon indirectly. These results paint a clear panorama of intestinal changes after PAT exposure and provide valuable information on the exposure risk and toxic mechanism of PAT.


Subject(s)
Metabolomics , Mice, Inbred C57BL , Patulin , Transcriptome , Animals , Patulin/toxicity , Mice , Transcriptome/drug effects , Male , Intestine, Small/drug effects , Intestine, Small/pathology , Intestine, Small/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Colon/drug effects , Colon/pathology , Intestines/drug effects , Intestines/pathology
3.
J Anim Sci ; 1022024 Jan 03.
Article in English | MEDLINE | ID: mdl-38619320

ABSTRACT

The present study aimed to investigate the effects of deoxynivalenol (DON) stimulation on inflammatory injury and the expression of the glucose transporters sodium-dependent glucose transporter 1 (SGLT1) and glucose transporter protein 2 (GLU2) in porcine small intestinal epithelial cells (IPEC-J2). Additionally, the study aimed to provide initial insights into the connection between the expression of glucose transporters and the inflammatory injury of IPEC-J2 cells. DON concentration and DON treatment time were determined using the CCK­8 assay. Accordingly, 1.0 µg/mL DON and treatment for 24 h were chosen for subsequent experiments. Then IPEC-J2 cells were treated without DON (CON, N = 6) or with 1 µg/mL DON (DON, N = 6). Lactate dehydrogenase (LDH) content, apoptosis rate, and proinflammatory cytokines including interleukin (IL)-1ß, Il-6, and tumor necrosis factor α (TNF-α) were measured. Additionally, the expression of AMP-activated protein kinase α1 (AMPK-α1), the content of glucose, intestinal alkaline phosphatase (AKP), and sodium/potassium-transporting adenosine triphosphatase (Na+/K+-ATPase) activity, and the expression of SGLT1 and GLU2 of IPEC-J2 cells were also analyzed. The results showed that DON exposure significantly increased LDH release and apoptosis rate of IPEC-J2 cells. Stimulation with DON resulted in significant cellular inflammatory damage, as evidenced by a significant increase in proinflammatory cytokines (IL-1ß, IL-6, and TNF-α). Additionally, DON caused damage to the glucose absorption capacity of IPEC-J2 cells, indicated by decreased levels of glucose content, AKP activity, Na+/K+-ATPase activity, AMPK-α1 protein expression, and SGLT1 expression. Correlation analysis revealed that glucose absorption capacity was negatively correlated with cell inflammatory cytokines. Based on the findings of this study, it can be preliminarily concluded that the cell inflammatory damage caused by DON may be associated with decreased glucose absorption.


Glucose is one of the most basic nutrients necessary to sustain animal life and plays a crucial role in animal body composition and energy metabolism. Previous studies suggested a link between glucose absorption and inflammatory injury. In the present study, deoxynivalenol (DON) stimulation caused severe inflammatory injury and reduced the glucose absorption capacity of IPEC-J2 cells. Pearson's correlation analysis revealed a negative correlation between glucose absorption capacity and cell inflammatory cytokines. Ultimately, it can be speculated that the cellular inflammatory response triggered by DON may be related to the altered expression of glucose transporters.


Subject(s)
Epithelial Cells , Glucose , Intestine, Small , Sodium-Glucose Transporter 1 , Trichothecenes , Animals , Trichothecenes/toxicity , Swine , Glucose/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 1/genetics , Cell Line , Intestine, Small/drug effects , Inflammation/chemically induced , Cytokines/metabolism , Cytokines/genetics , Biological Transport/drug effects , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 2/genetics , Apoptosis/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism
4.
Sci Rep ; 14(1): 8905, 2024 04 17.
Article in English | MEDLINE | ID: mdl-38632282

ABSTRACT

Glyphosate is the active ingredient of glyphosate-based herbicides and the most commonly used pesticide in the world. The goal of the present study was to verify whether low doses of glyphosate (equivalent to the environmental exposure) evoke changes in galanin expression in intramural neurons in the small intestine in pigs and to quantitatively determine changes in the level of galanin receptor encoding mRNA (GALR1, GALR2, GALR3) in the small intestine wall. The experiment was conducted on 15 sexually immature gilts divided into three study groups: control (C)-animals receiving empty gelatin capsules; experimental 1 (G1)-animals receiving a low dose of glyphosate (0.05 mg/kg b.w./day); experimental 2 (G2)-animals receiving a higher dose of glyphosate (0.5 mg/kg b.w./day) orally in gelatine capsules for 28 days. Glyphosate ingestion led to an increase in the number of GAL-like immunoreactive intramural neurons in the porcine small intestine. The results of RT-PCR showed a significant increase in the expression of mRNA, which encodes the GAL-receptors in the ileum, a decreased expression in the duodenum and no significant changes in the jejunum. Additionally, intoxication with glyphosate increased the expression of SOD2-encoding mRNA in the duodenum and decreased it in the jejunum and ileum, but it did not affect SOD1 expression. The results suggest that it may be a consequence of the cytotoxic and/or neurotoxic properties of glyphosate and/or its ability to induce oxidative stress.


Subject(s)
Galanin , Glyphosate , Animals , Female , Galanin/metabolism , Glyphosate/metabolism , Glyphosate/toxicity , Intestine, Small/drug effects , Intestine, Small/metabolism , Receptor, Galanin, Type 2/drug effects , Receptor, Galanin, Type 2/genetics , Receptor, Galanin, Type 2/metabolism , RNA, Messenger/metabolism , Sus scrofa/genetics , Swine , Receptor, Galanin, Type 1/drug effects , Receptor, Galanin, Type 1/genetics , Receptor, Galanin, Type 1/metabolism , Receptor, Galanin, Type 3/drug effects , Receptor, Galanin, Type 3/genetics , Receptor, Galanin, Type 3/metabolism , Herbicides/toxicity
5.
J Proteome Res ; 23(5): 1801-1809, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38655769

ABSTRACT

Alcohol consumption perturbs the gut immune barrier and ultimately results in alcoholic liver diseases, but little is known about how immune-related cells in the gut are perturbed in this process. In this study, we employed laser capture microdissection and a label-free proteomics approach to investigate the consequences of alcohol exposure to the proteomes of crypts and villi in the proximal small intestine. Intestinal tissues from alcohol-fed and pair-fed mice were microdissected to selectively capture cells in the crypts and villi regions, followed by one-pot protein digestion and data-independent LC-MS/MS analysis. We successfully identified over 3000 proteins from each of the crypt or villi regions equivalent to ∼3000 cells. Analysis of alcohol-treated tissues indicated an enhanced alcohol metabolism and reduced levels of α-defensins in crypts, alongside increased lipid metabolism and apoptosis in villi. Immunofluorescence imaging further corroborated the proteomic findings. Our work provides a detailed profiling of the proteomic changes in the compartments of the mouse small intestine and aids in molecular-level understanding of alcohol-induced tissue damage.


Subject(s)
Ethanol , Intestine, Small , Proteomics , Animals , Intestine, Small/metabolism , Intestine, Small/drug effects , Intestine, Small/pathology , Proteomics/methods , Mice , Ethanol/toxicity , Tandem Mass Spectrometry , Proteome/metabolism , Proteome/analysis , Proteome/drug effects , Laser Capture Microdissection , Chromatography, Liquid , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Mice, Inbred C57BL , Male , Apoptosis/drug effects , Lipid Metabolism/drug effects
6.
Mol Nutr Food Res ; 68(9): e2300889, 2024 May.
Article in English | MEDLINE | ID: mdl-38676468

ABSTRACT

SCOPE: Epidemiological studies have linked excessive red and processed meat intake to gut disorders. Under laboratory conditions, high heme content is considered the primary health risk factor for red meat. However, heme in meat is present in myoglobin, which is an indigestible protein, suggesting the different functions between myoglobin and heme. This study aims to explore how dietary myoglobin and heme affect gut health and microbiota differently. METHODS AND RESULTS: Histological and biochemical assessments as well as 16S rRNA sequencing are performed. Moderate myoglobin intake (equivalent to the recommended intake of 150 g meat per day for human) has beneficial effects on the duodenal barrier. However, a too high myoglobin diet (equivalent to intake of 3000 g meat per day for human) triggers duodenum injury and alters the microbial community. The hemin diet destroys intestinal tissue and ileal microbiota more significantly. The in vitro experiments further confirm that free heme exhibits high toxicity to beneficial gut bacteria while myoglobin promotes the growth and metabolism of Limosilactobacillus reuteri. CONCLUSION: Moderate intake of myoglobin or hemin is beneficial to intestinal health and microbiota, but too high amounts lead to tissue inflammation and injury in the small intestine by reshaping ileal microbiota.


Subject(s)
Gastrointestinal Microbiome , Hemin , Inflammation , Myoglobin , Gastrointestinal Microbiome/drug effects , Animals , Myoglobin/metabolism , Hemin/pharmacology , Male , Diet/methods , Intestine, Small/drug effects , Intestine, Small/metabolism , Limosilactobacillus reuteri , Duodenum/metabolism , RNA, Ribosomal, 16S/genetics , Heme
7.
Phytomedicine ; 128: 155363, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38493715

ABSTRACT

BACKGROUND: Coccidiosis is a rapidly spreading and acute parasitic disease that seriously threatening the intestinal health of poultry. Matrine from leguminous plants has anthelmintic and anti-inflammatory properties. PURPOSE: This assay was conducted to explore the protective effects of Matrine and the AntiC (a Matrine compound) on Eimeria necatrix (EN)-infected chick small intestines and to provide a nutritional intervention strategy for EN injury. STUDY DESIGN: The in vivo (chick) experiment: A total of 392 one-day-old yellow-feathered broilers were randomly assigned to six groups in a 21-day study: control group, 350 mg/kg Matrine group, 500 mg/kg AntiC group, EN group, and EN + 350 mg/kg Matrine group, EN + 500 mg/kg AntiC group. The in vitro (chick intestinal organoids, IOs): The IOs were treated with PBS, Matrine, AntiC, 3 µM CHIR99021, EN (15,000 EN sporozoites), EN + Matrine, EN + AntiC, EN + Matrine + CHIR99021, EN + AntiC + CHIR99021. METHODS: The structural integrity of chicks jejunal crypt-villus axis was evaluated by hematoxylin and eosin (H&E) staining and transmission electron microscopy (TEM). And the activity of intestinal stem cells (ISCs) located in crypts was assessed by in vitro expansion advantages of a primary in IOs model. Then, the changes of Wnt/ß-catenin signaling in jejunal tissues and IOs were detected by Real-Time qPCR,Western blotting and immunohistochemistry. RESULTS: The results showed that dietary supplementation with Matrine or AntiC rescued the jejunal injury caused by EN, as indicated by increased villus height, reduced crypt hyperplasia, and enhanced expression of tight junction proteins. Moreover, there was less budding efficiency of the IOs expanded from jejunal crypts of chicks in the EN group than that in the Matrine and AntiC group, respectively. Further investigation showed that AntiC and Matrine inhibited EN-stimulated Wnt/ß-catenin signaling. The fact that Wnt/ß-catenin activation via CHIR99021 led to the failure of Matrine and AntiC to rescue damaged ISCs confirmed the dominance of this signaling. CONCLUSION: Our results suggest that Matrine and AntiC inhibit ISC proliferation and promote ISC differentiation into absorptive cells by preventing the hyperactivation of Wnt/ß-catenin signaling, thereby standardizing the function of ISC proliferation and differentiation, which provides new insights into mitigating EN injury by Matrine and AntiC.


Subject(s)
Alkaloids , Chickens , Coccidiosis , Eimeria , Matrines , Poultry Diseases , Quinolizines , Wnt Signaling Pathway , Animals , Quinolizines/pharmacology , Alkaloids/pharmacology , Wnt Signaling Pathway/drug effects , Eimeria/drug effects , Coccidiosis/drug therapy , Poultry Diseases/drug therapy , Poultry Diseases/parasitology , Stem Cells/drug effects , Intestine, Small/drug effects , Intestine, Small/parasitology
8.
Biomater Sci ; 12(9): 2312-2320, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38497434

ABSTRACT

Postsurgical treatment comprehensively benefits from the application of tissue-adhesive injectable hydrogels, which reduce postoperative complications by promoting wound closure and tissue regeneration. Although various hydrogels have been employed as clinical tissue adhesives, many exhibit deficiencies in adhesive strength under wet conditions or in immunomodulatory functions. Herein, we report the development of reactive oxygen species (ROS) scavenging and tissue-adhesive injectable hydrogels composed of polyamine-modified gelatin crosslinked with the 4-arm poly (ethylene glycol) crosslinker. Polyamine-modified gelatin was particularly potent in suppressing the secretion of proinflammatory cytokines from stimulated primary macrophages. This effect is attributed to its ability to scavenge ROS and inhibit the nuclear translocation of nuclear factor kappa-B. Polyamine-modified gelatin-based hydrogels exhibited ROS scavenging abilities and enhanced tissue adhesive strength on collagen casing. Notably, the hydrogel demonstrated exceptional tissue adhesive properties in a wet environment, as evidenced by its performance using porcine small intestine tissue. This approach holds significant promise for designing immunomodulatory hydrogels with superior tissue adhesion strength compared to conventional medical materials, thereby contributing to advancements in minimally invasive surgical techniques.


Subject(s)
Gelatin , Hydrogels , Reactive Oxygen Species , Tissue Adhesives , Hydrogels/chemistry , Hydrogels/administration & dosage , Hydrogels/pharmacology , Animals , Tissue Adhesives/chemistry , Tissue Adhesives/pharmacology , Tissue Adhesives/administration & dosage , Reactive Oxygen Species/metabolism , Mice , Swine , Gelatin/chemistry , Polyethyleneimine/chemistry , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Free Radical Scavengers/administration & dosage , Macrophages/drug effects , Macrophages/metabolism , Polyethylene Glycols/chemistry , Injections , Cytokines/metabolism , Intestine, Small/metabolism , Intestine, Small/drug effects
9.
Poult Sci ; 103(5): 103582, 2024 May.
Article in English | MEDLINE | ID: mdl-38457989

ABSTRACT

Small peptides are nutrients and bioactive molecules that have dual regulatory effects on nutrition and physiology. They are of great significance for maintaining the intestinal health and production performance of broilers. We here cultured the primary small intestinal epithelial cells (IEC) of chicken in a medium containing L-Leu (Leu) and L-Leu-L-Leu (Leu-Leu) for 24 h. The untreated cells were considered as the control group. The growth, proliferation, and apoptosis of IEC were examined. By combining RNA-seq and label-free sequencing technology, candidate genes, proteins, and pathways related to the growth, proliferation, and apoptosis of IEC were screened. Immunofluorescence detection revealed that the purity of the isolated primary IEC was >90%. The Leu-Leu group significantly promoted IEC growth and proliferation and significantly inhibited IEC apoptosis, and the effect was better than those of the Leu and control groups. Using transcriptome sequencing, four candidate genes, CCL20, IL8L1, IL8, and IL6, were screened in the Leu group, and one candidate gene, IL8, was screened in the Leu-Leu group. Two candidate genes, IL6 and RGN, were screened in the Leu-Leu group compared with the Leu group. Nonquantitative proteomic marker sequencing results revealed that through the screening of candidate proteins and pathways, found one growth-related candidate protein PGM3 and three proliferation-related candidate proteins RPS17, RPS11, and RPL23, and two apoptosis-related candidate proteins GPX4 and PDPK1 were found in the Leu-Leu group compared with Leu group. In short, Leu-Leu could promote IEC growth and proliferation and inhibit IEC apoptosis. On combining transcriptome and proteome sequencing technologies, multiple immune- and energy-related regulatory signal pathways were found to be related to IEC growth, proliferation, and apoptosis. Three candidate genes of IL8, IL6, and RGN were identified, and six candidate proteins of PGM3, RPS17, RPS11, RPL23, GPX4, and PDPK1 were involved in IEC growth, proliferation, and apoptosis. The results provide valuable data for preliminarily elucidating small peptide-mediated IEC regulation pathways, improving the small peptide nutrition theoretical system, and establishing small peptide nutrition regulation technology.


Subject(s)
Apoptosis , Cell Proliferation , Chickens , Epithelial Cells , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/physiology , Intestinal Mucosa/drug effects , Intestinal Mucosa/cytology , Intestine, Small/cytology , Intestine, Small/drug effects , Avian Proteins/genetics , Avian Proteins/metabolism
10.
Nature ; 621(7977): 179-187, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37648857

ABSTRACT

Tissue resident memory CD8+ T (TRM) cells offer rapid and long-term protection at sites of reinfection1. Tumour-infiltrating lymphocytes with characteristics of TRM cells maintain enhanced effector functions, predict responses to immunotherapy and accompany better prognoses2,3. Thus, an improved understanding of the metabolic strategies that enable tissue residency by T cells could inform new approaches to empower immune responses in tissues and solid tumours. Here, to systematically define the basis for the metabolic reprogramming supporting TRM cell differentiation, survival and function, we leveraged in vivo functional genomics, untargeted metabolomics and transcriptomics of virus-specific memory CD8+ T cell populations. We found that memory CD8+ T cells deployed a range of adaptations to tissue residency, including reliance on non-steroidal products of the mevalonate-cholesterol pathway, such as coenzyme Q, driven by increased activity of the transcription factor SREBP2. This metabolic adaptation was most pronounced in the small intestine, where TRM cells interface with dietary cholesterol and maintain a heightened state of activation4, and was shared by functional tumour-infiltrating lymphocytes in diverse tumour types in mice and humans. Enforcing synthesis of coenzyme Q through deletion of Fdft1 or overexpression of PDSS2 promoted mitochondrial respiration, memory T cell formation following viral infection and enhanced antitumour immunity. In sum, through a systematic exploration of TRM cell metabolism, we reveal how these programs can be leveraged to fuel memory CD8+ T cell formation in the context of acute infections and enhance antitumour immunity.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocytes, Tumor-Infiltrating , Neoplasms , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Respiration , Cholesterol/metabolism , Cholesterol/pharmacology , Immunologic Memory , Intestine, Small/drug effects , Intestine, Small/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Metabolomics , Mevalonic Acid/metabolism , Neoplasms/immunology , Ubiquinone/metabolism , Virus Diseases/immunology , Viruses/immunology , Mitochondria/metabolism
11.
Int. j. morphol ; 40(6): 1466-1474, dic. 2022. ilus, tab
Article in English | LILACS | ID: biblio-1421816

ABSTRACT

SUMMARY: Fifty male Wistar albino rats were divided into 5 groups; Group 1 as a sham group. Group 2 as a control group, Group 3 as 100 mg/kg CDP-choline administered group, Group as 200 mg/kg CDP-choline administered group, and Group 5 as sepsis group. The sepsis model was performed by ligating and perforating the caecum of rats. Liver and small intestine tissues were assessed either histologically or quantitatively and qualitatively. There was a significant difference between the sepsis and CDP-choline groups for liver and intestinal damage evaluated in tissue samples. (p <0.001). CDP-choline treatment partially improved dose-dependent the clinical parameters of sepsis and septic shock, reversed micro-anatomical damage caused by sepsis.


Cincuenta ratas albinas Wistar macho se dividieron en 5 grupos; Grupo 1 como grupo control simulador, el grupo 2 como grupo de control, el grupo 3 como grupo al que se administró 100 mg/kg de CDP-colina, el grupo 4 como grupo al que se administró 200 mg/kg de CDP-colina y el grupo 5 como grupo con sepsis. El modelo de sepsis se realizó ligando y perforando el intestino ciego de las ratas. Los tejidos del hígado y del intestino delgado se evaluaron histológicamente o cuantitativa y cualitativamente. Hubo una diferencia significativa entre los grupos de sepsis y CDP-colina para el daño hepático e intestinal evaluado en muestras de tejido (p<0,001). El tratamiento con CDP-colina mejoró parcialmente, según la dosis, los parámetros clínicos de sepsis y shock séptico y revirtió el daño micro anatómico causado por la sepsis.


Subject(s)
Animals , Rats , Sepsis/drug therapy , Cytidine Diphosphate Choline/administration & dosage , Intestine, Small/drug effects , Liver/drug effects , Rats, Wistar , Cytidine Diphosphate Choline/pharmacology , Disease Models, Animal , Intestine, Small/pathology , Liver/pathology
12.
Int J Mol Sci ; 23(3)2022 Jan 24.
Article in English | MEDLINE | ID: mdl-35163210

ABSTRACT

Doxorubicin is widely used in the treatment of different cancers, and its side effects can be severe in many tissues, including the intestines. Symptoms such as diarrhoea and abdominal pain caused by intestinal inflammation lead to the interruption of chemotherapy. Nevertheless, the molecular mechanisms associated with doxorubicin intestinal toxicity have been poorly explored. This study aims to investigate such mechanisms by exposing 3D small intestine and colon organoids to doxorubicin and to evaluate transcriptomic responses in relation to viability and apoptosis as physiological endpoints. The in vitro concentrations and dosing regimens of doxorubicin were selected based on physiologically based pharmacokinetic model simulations of treatment regimens recommended for cancer patients. Cytotoxicity and cell morphology were evaluated as well as gene expression and biological pathways affected by doxorubicin. In both types of organoids, cell cycle, the p53 signalling pathway, and oxidative stress were the most affected pathways. However, significant differences between colon and SI organoids were evident, particularly in essential metabolic pathways. Short time-series expression miner was used to further explore temporal changes in gene profiles, which identified distinct tissue responses. Finally, in silico proteomics revealed important proteins involved in doxorubicin metabolism and cellular processes that were in line with the transcriptomic responses, including cell cycle and senescence, transport of molecules, and mitochondria impairment. This study provides new insight into doxorubicin-induced effects on the gene expression levels in the intestines. Currently, we are exploring the potential use of these data in establishing quantitative systems toxicology models for the prediction of drug-induced gastrointestinal toxicity.


Subject(s)
Doxorubicin/toxicity , Intestines/drug effects , Intestines/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Colon/drug effects , Doxorubicin/pharmacology , Gene Expression/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Humans , Intestine, Small/drug effects , Models, Biological , Organoids/cytology , Organoids/drug effects , Organoids/metabolism , Proteomics , Transcriptome/genetics
13.
Biochem Biophys Res Commun ; 595: 7-13, 2022 03 05.
Article in English | MEDLINE | ID: mdl-35091109

ABSTRACT

The intestinal tract is an essential component of the body's immune system, and is extremely sensitive to exposure of ionizing radiation. While ionizing radiation can effectively induce multiple forms of cell death, whether it can also promote ferroptosis in intestinal cells and the possible interrelationship between ferroptosis and intestinal immune function has not been reported so far. Here, we found that radiation-induced major ultrastructural changes in mitochondria of small intestinal epithelial cells and the changes induced in iron content and MDA levels in the small intestine were consistent with that observed during cellular ferroptosis, thus suggesting occurrence of ferroptosis in radiation-induced intestinal damage. Moreover, radiation caused a substantial increase in the expression of ferroptosis-related factors such as LPCAT3 and ALOX15 mRNA, augmented the levels of immune-related factors INF-γ and TGF-ß mRNA, and decreased the levels of IL-17 mRNA thereby indicating that ionizing radiation induced ferroptosis and impairment of intestinal immune function. Liproxstatin-1 is a ferroptosis inhibitor that was found to ameliorate radiation-induced ferroptosis and promote the recovery from immune imbalances. These findings supported the role of ferroptosis in radiation-induced intestinal immune injury and provide novel strategies for protection against radiation injury through regulation of the ferroptosis pathway.


Subject(s)
Ferroptosis/physiology , Intestines/pathology , Quinoxalines/pharmacology , Radiation Injuries, Experimental/prevention & control , Radiation, Ionizing , Spiro Compounds/pharmacology , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/metabolism , Ferroptosis/drug effects , Ferroptosis/radiation effects , Gene Expression/drug effects , Gene Expression/radiation effects , Glutathione/metabolism , Intestine, Small/drug effects , Intestine, Small/metabolism , Intestine, Small/radiation effects , Intestines/drug effects , Intestines/radiation effects , Male , Malondialdehyde/metabolism , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/radiation effects , Mitochondria/ultrastructure , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/physiopathology , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/metabolism
14.
Arch Toxicol ; 96(2): 499-510, 2022 02.
Article in English | MEDLINE | ID: mdl-34654938

ABSTRACT

The small intestine plays a critical role in the absorption and metabolism of orally administered drugs. Therefore, a model capable of evaluating drug absorption and metabolism in the small intestine would be useful for drug discovery. Patients with genotype UGT1A1*6 (exon 1, 211G > A) treated with the antineoplastic drug SN-38 have been reported to exhibit decreased glucuronide conjugation and increased incidence of intestinal toxicity and its severe side effects, including severe diarrhea. To ensure the safety of drugs, we must develop a drug metabolism and toxicity evaluation model which considers UGT1A1*6. In this study, we generated CYP3A4·POR·UGT1A1 KI- and CYP3A4·POR·UGT1A1*6 KI-Caco-2 cells for pharmaceutical research using a PITCh system. The CYP3A4·POR·UGT1A1 KI-Caco-2 cells were shown to express functional CYP3A4 and UGT1A1. The CYP3A4·POR·UGT1A1*6 KI-Caco-2 cells were sensitive to SN-38-induced intestinal toxicity. We thus succeeded in generating CYP3A4·POR·UGT1A1 KI- and CYP3A4·POR·UGT1A1*6 KI-Caco-2 cells, which can be used in pharmaceutical research. We also developed an intestinal epithelial cell model of patients with UGT1A1*6 and showed that it was useful as a tool for drug discovery.


Subject(s)
Cytochrome P-450 CYP3A/genetics , Glucuronosyltransferase/genetics , Intestinal Mucosa/enzymology , Intestine, Small/enzymology , Antineoplastic Agents/toxicity , Caco-2 Cells/enzymology , Drug Discovery/methods , Genotype , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestine, Small/cytology , Intestine, Small/drug effects , Irinotecan/toxicity
15.
J Neuroimmunol ; 362: 577764, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34823118

ABSTRACT

Muscarinic-acetylcholine-receptors (mAChRs) modulate intestinal homeostasis, but their role in inflammation is unclear; thus, this issue was the focus of this study. BALB/c mice were treated for 7 days with muscarine (mAChR/agonist), atropine (mAChR/antagonist) or saline. Small-intestine samples were collected for histology and cytofluorometric assays in Peyer's patches (PP) and lamina propria (LP) cell-suspensions. In LP, goblet-cells/leukocytes/neutrophils/MPO+ cells and MPO/activity were increased in the muscarine group. In PP, IFN-γ+/CD4+ T or IL-6+/CD4+ T cell numbers were higher in the muscarine or atropine groups, respectively. In LP, TNF-α+/CD4+ T cell number was higher in the muscarine group and lower in the atropine.


Subject(s)
Inflammation/immunology , Intestinal Mucosa/immunology , Receptors, Muscarinic/immunology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Intestinal Mucosa/drug effects , Intestine, Small/drug effects , Intestine, Small/immunology , Mice , Mice, Inbred BALB C , Muscarinic Agonists/pharmacology , Peyer's Patches/drug effects , Peyer's Patches/immunology
16.
Biomed Pharmacother ; 146: 112496, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34959117

ABSTRACT

Intestinal injury is one of the major side effects that are induced by medical radiation exposure, and has limited effective therapies. In this study, we investigated the beneficial effects of sanguinarine (SAN) on intestinal injury induced by ionizing radiation (IR) both in vitro and in vivo. Mice were exposed to whole abdominal irradiation (WAI) to mimic clinical scenarios. SAN was injected intraperitoneally to mitigate IR-induced injury. Histological examination was performed to assess the tissue injuries of the spleen and small intestine. A small intestinal epithelial cell line-6 (IEC-6) was analyzed for its viability and apoptosis in vitro under different treatments. Inflammation-related pathways and serum inflammatory cytokines were detected via Western blot analysis and ELISA, respectively. High-throughput sequencing was used to characterize the gut microbiota profile. High-performance liquid chromatography was performed to assess short-chain fatty acid contents in the colon. In vitro, SAN pretreatment protected cell viability and reduced apoptosis in IEC-6 cells. In vivo, SAN pretreatment protected immune organs, alleviated intestinal injury, and promoted intestinal recovery. SAN also reduced the levels of inflammatory cytokines, suppressed high mobility group box 1 (HMGB1)/ Toll-like receptor 4 (TLR4) pathway activation, and modulated gut microbiota composition. Our findings demonstrate that the beneficial properties of SAN alleviated intestinal radiation injury. Thus, SAN represents a therapeutic option for protecting against IR-induced intestinal injury in preclinical settings.


Subject(s)
Benzophenanthridines/pharmacology , Intestine, Small/drug effects , Isoquinolines/pharmacology , Radiation Injuries, Experimental/prevention & control , Spleen/drug effects , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Cytokines/drug effects , Dose-Response Relationship, Drug , Down-Regulation , Fatty Acids, Volatile , Gastrointestinal Microbiome/drug effects , HMGB1 Protein/drug effects , Inflammation Mediators/metabolism , Injections, Intraperitoneal , Intestine, Small/pathology , Intestine, Small/radiation effects , Male , Mice , Mice, Inbred C57BL , Radiation Injuries, Experimental/pathology , Radiation, Ionizing , Signal Transduction/drug effects , Spleen/pathology , Spleen/radiation effects , Toll-Like Receptor 4/drug effects
17.
BMC Cancer ; 21(1): 1343, 2021 Dec 18.
Article in English | MEDLINE | ID: mdl-34922485

ABSTRACT

BACKGROUND: Chemotherapy is frequently used in cancer treatment; however, it may cause adverse events, which must be managed. Reactive oxygen species (ROS) have been reported to be involved in the induction of intestinal mucositis and diarrhea, which are common side effects of treatment with fluoropyrimidine 5-fluorouracil (5-FU). Our previous studies have shown that oral administration of cystine and theanine (CT) increases glutathione (GSH) production in vivo. In the present study, we hypothesized that CT might inhibit oxidative stress, including the overproduction of ROS, and attenuate 5-FU-induced mucositis and diarrhea. METHODS: We investigated the inhibitory effect of CT administration on mucositis and diarrhea, as well as its mechanism, using a mouse model of 5-FU-induced intestinal mucositis. RESULTS: CT administration suppressed 5-FU-induced diarrhea and weight loss in the studied mice. After 5-FU administration, the GSH level and the GSH/GSSG ratio in the small intestine mucosal tissue decreased compared to normal control group; but CT administration improved the GSH/GSSG ratio to normal control levels. 5-FU induced ROS production in the basal region of the crypt of the small intestine mucosal tissue, which was inhibited by CT. CT did not affect the antitumor effect of 5-FU. CONCLUSIONS: CT administration suppressed intestinal mucositis and diarrhea in a mouse model. This finding might be associated with the antioxidant characteristics of CT, including the improved rate of GSH redox and the reduced rate of ROS production in the small intestine mucosal tissue. CT might be a suitable candidate for the treatment of gastrointestinal mucositis associated with chemotherapy.


Subject(s)
Cystine/administration & dosage , Diarrhea/drug therapy , Fluorouracil/adverse effects , Glutamates/administration & dosage , Mucositis/drug therapy , Animals , Diarrhea/chemically induced , Diarrhea/immunology , Diarrhea/pathology , Disease Models, Animal , Drug Therapy, Combination/methods , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestine, Small/drug effects , Intestine, Small/immunology , Intestine, Small/pathology , Male , Mice , Mucositis/chemically induced , Mucositis/immunology , Mucositis/pathology , Oxidative Stress/drug effects , Oxidative Stress/immunology , Reactive Oxygen Species/metabolism
18.
Oxid Med Cell Longev ; 2021: 9927864, 2021.
Article in English | MEDLINE | ID: mdl-34795844

ABSTRACT

Bisdemethoxycurcumin is one of the three curcuminoids of turmeric and exhibits good antioxidant activity in animal models. This study is aimed at investigating the effect of bisdemethoxycurcumin on small intestinal mitochondrial dysfunction in lipopolysaccharide- (LPS-) treated broilers, especially on the mitochondrial thioredoxin 2 system and mitochondrial biogenesis. A total of 320 broiler chickens were randomly assigned into four experimental diets using a 2 × 2 factorial arrangement with diet (0 and 150 mg/kg bisdemethoxycurcumin supplementation) and stress (saline or LPS challenge) for 20 days. Broilers received a dose of LPS (1 mg/kg body weight) or sterile saline intraperitoneally on days 16, 18, and 20 of the trial. Bisdemethoxycurcumin mitigated the mitochondrial dysfunction of jejunum and ileum induced by LPS, as evident by the reduced reactive oxygen species levels and the increased mitochondrial membrane potential. Bisdemethoxycurcumin partially reversed the decrease in the mitochondrial DNA copy number and the depletion of ATP levels. Bisdemethoxycurcumin activated the mitochondrial antioxidant response, including the prevention of lipid peroxidation, enhancement of manganese superoxide dismutase activity, and the upregulation of the mitochondrial glutaredoxin 5 and thioredoxin 2 system. The enhanced mitochondrial respiratory complex activities in jejunum and ileum were also attributed to bisdemethoxycurcumin treatment. In addition, bisdemethoxycurcumin induced mitochondrial biogenesis via transcriptional regulation of proliferator-activated receptor-gamma coactivator-1alpha pathway. In conclusion, our results demonstrated the potential of bisdemethoxycurcumin to attenuate small intestinal mitochondrial dysfunction, which might be mediated via activating the mitochondrial antioxidant system and mitochondrial biogenesis in LPS-treated broilers.


Subject(s)
Antioxidants/metabolism , Diarylheptanoids/pharmacology , Intestinal Diseases/prevention & control , Intestine, Small/drug effects , Lipopolysaccharides/toxicity , Mitochondria/drug effects , Protective Agents/pharmacology , Animals , Chickens , Gene Expression Regulation , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Intestinal Diseases/chemically induced , Intestinal Diseases/metabolism , Intestinal Diseases/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Mitochondria/metabolism , Mitochondria/pathology , Reactive Oxygen Species/metabolism
19.
Sci Rep ; 11(1): 22733, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34815475

ABSTRACT

Small intestinal neuroendocrine tumors (SI-NETs) are slow-growing tumors that seem genetically quite stable without highly recurrent mutations, but are epigenetically dysregulated. In contrast to the undetectable expression of the enhancer of zeste homolog 2 (EZH2) histone methyltransferase in the enterochromaffin cells of the small intestine, we found high and differential expression of EZH2 in primary SI-NETs and corresponding metastases. Silencing EZH2 in the SI-NET cell line CNDT2.5 reduced cell proliferation and induced apoptosis. Furthermore, EZH2 knockout inhibited tumor progression in a CNDT2.5 SI-NET xenograft mouse model, and treatment of SI-NET cell lines CNDT2.5 and GOT1 with the EZH2-specific inhibitor CPI-1205 decreased cell viability and promoted apoptosis. Moreover, CPI-1205 treatment reduced migration capacity of CNDT2.5 cells. The EZH2 inhibitor GSK126 also repressed proliferation of CNDT2.5 cells. Recently, metformin has received wide attention as a therapeutic option in diverse cancers. In CNDT2.5 and GOT1 cells, metformin suppressed EZH2 expression, and inhibited cell proliferation. Exposure of GOT1 three-dimensional cell spheroids to CPI-1205 or metformin arrested cell proliferation and decreased spheroid size. These novel findings support a possible role of EZH2 as a candidate oncogene in SI-NETs, and suggest that CPI-1205 and metformin should be further evaluated as therapeutic options for patients with SI-NETs.


Subject(s)
Biomarkers, Tumor/metabolism , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Indoles/pharmacology , Intestinal Neoplasms/drug therapy , Intestine, Small/drug effects , Neuroendocrine Tumors/drug therapy , Piperidines/pharmacology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Drug Therapy, Combination , Enzyme Inhibitors/pharmacology , Female , Humans , Hypoglycemic Agents/pharmacology , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Intestine, Small/pathology , Metformin/pharmacology , Mice , Mice, Nude , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
Biol Pharm Bull ; 44(11): 1746-1751, 2021.
Article in English | MEDLINE | ID: mdl-34719650

ABSTRACT

Tramadol is a weak opioid that produces analgesic effect via both the µ-opioid receptor (MOR) and non-opioid targets. Constipation is the most common opioid-related side effect in patients with cancer and non-cancer pain. However, the contribution of MOR to tramadol-induced constipation is unclear. Therefore, we used naldemedine, a peripherally acting MOR antagonist, and MOR-knockout mice to investigate the involvement of peripheral MOR in tramadol-induced constipation using a small intestinal transit model. A single dose of tramadol (3-100 mg/kg, per os (p.o.)) inhibited small intestinal transit dose-dependently in rats. Naldemedine (0.01-10 mg/kg, p.o.) blocked the inhibition of small intestinal transit induced by tramadol (30 mg/kg, p.o.) in rats. The transition rate increased dose-dependently over the range of naldemedine 0.01-0.3 mg/kg, and complete recovery was observed at 0.3-10 m/kg. Additionally, tramadol (30 and 100 mg/kg, subcutaneously (s.c.)) inhibited small intestinal transit in wild-type mice but not in MOR-knockout mice. These results suggest that peripheral MOR participates in tramadol-induced constipation.


Subject(s)
Analgesics, Opioid/adverse effects , Opioid-Induced Constipation/etiology , Receptors, Opioid, mu/drug effects , Tramadol/adverse effects , Analgesics, Opioid/blood , Analgesics, Opioid/pharmacokinetics , Animals , Intestine, Small/drug effects , Male , Naltrexone/adverse effects , Naltrexone/analogs & derivatives , Naltrexone/blood , Naltrexone/pharmacokinetics , Nociception/drug effects , Opioid-Induced Constipation/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Opioid, mu/metabolism , Tramadol/blood , Tramadol/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...