Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 949
Filtrar
1.
Narra J ; 4(2): e898, 2024 08.
Artigo em Inglês | MEDLINE | ID: mdl-39280279

RESUMO

Enteropathy is a gastrointestinal disorder characterized by inflammation in the small intestine and one of the causes of enteropathy is the side effects of certain drugs, such as non-steroidal anti-inflammatory drugs (NSAIDs). The mechanism of NSAIDs, such as indomethacin, could inhibit prostaglandin synthesis, leading to a decrease in mucus production and small intestine integrity. To test the effects of a drug, it is necessary to undergo preclinical testing using animal models. Commonly used animal models such as mice and rats have several drawbacks including high cost, ethical issues, and long lifespan. Therefore, alternatives such as using invertebrate animals like Drosophila melanogaster as a more economical in vivo platform with genetic similarity to mammals and devoid of ethical concerns are needed. The aim of this study was to evaluate Drosophila melanogaster as an in vivo model organism in testing the side effects of pharmaceuticals that cause enteropathy. In this study, flies aged 3-5 days were starved and then placed into treatment vials comprising untreated control and indomethacin-treated (3.75 mM, 7.5 mM, and 15 mM). Survival analysis was conducted during the treatment period, followed by a Smurf assay test after seven days of treatment. Subsequently, the expression of pro-inflammatory cytokine-related genes (drs and totA), mitochondria stability-related genes (tom40), and endogenous antioxidant-related genes (sod1, sod2, and cat) was performed using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Our data indicated that indomethacin did not impact lifespan or cause intestinal damage. However, we observed increased expression of pro-inflammatory cytokine-related genes, including drs, and a twofold increase in totA gene expression. Furthermore, there was a significant upregulation of mitochondrial stability gene tom40, endogenous antioxidant genes sod1 and cat, and a threefold increase in sod2 at 15 mM indomethacin. Although no phenotypical changes in gut integrity were detected, the increased expression of pro-inflammatory cytokine genes suggests the occurrence of inflammation in the indomethacin-treated flies.


Assuntos
Anti-Inflamatórios não Esteroides , Drosophila melanogaster , Indometacina , Enteropatias , Animais , Drosophila melanogaster/efeitos dos fármacos , Indometacina/efeitos adversos , Indometacina/farmacologia , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/efeitos adversos , Enteropatias/induzido quimicamente , Enteropatias/patologia , Enteropatias/genética , Enteropatias/tratamento farmacológico , Modelos Animais de Doenças , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo
2.
Rev Med Suisse ; 20(887): 1665-1668, 2024 Sep 18.
Artigo em Francês | MEDLINE | ID: mdl-39295263

RESUMO

Olmesartan-induced enteropathy was first described twelve years ago. Clinically it is characterized by diarrhea, weight loss and malabsorption. Histological analysis may show duodenal villous atrophy and/or epithelial lymphocytosis (duodenal/colic). Celiac-specific antibodies are negative and gluten avoidance does not improve the symptomatology. This adverse event can occur months or years after the introduction of the causative drug, making it a real diagnostic challenge. The treatment is the avoidance of olmesartan, which will lead to both clinical and histological improvement.


L'entéropathie induite par l'olmésartan est une entité connue depuis une dizaine d'années et se caractérise par un syndrome de malabsorption avec diarrhées et perte pondérale. L'analyse histologique peut montrer une atrophie villositaire duodénale et/ou une lymphocytose épithéliale (duodénale et colique). Les anticorps spécifiques de la maladie cœliaque sont négatifs et l'éviction du gluten n'améliore en rien la symptomatologie. Cet effet indésirable peut se manifester des mois voire des années après l'introduction du traitement, ce qui en fait un réel défi diagnostique. Le traitement est l'éviction de la molécule, accompagnée d'une amélioration tant sur le plan clinique qu'histologique.


Assuntos
Imidazóis , Humanos , Imidazóis/efeitos adversos , Imidazóis/administração & dosagem , Tetrazóis/efeitos adversos , Bloqueadores do Receptor Tipo 1 de Angiotensina II/efeitos adversos , Enteropatias/induzido quimicamente , Enteropatias/diagnóstico , Diarreia/induzido quimicamente , Redução de Peso
3.
J Nutr Biochem ; 133: 109719, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39103108

RESUMO

This study investigated the protective effect of dulcitol on LPS-induced intestinal injury in piglets and explored the underlying molecular mechanisms. A total of 108 piglets were divided into three groups: CON, LPS, and DUL. The CON and LPS groups were fed a basal diet, the DUL group was fed a diet supplementation with 500 mg/kg dulcitol. On day 29, 6 piglets in the LPS and DUL groups were injected with 100 µg/kg BW of LPS. At 4 h postchallenge, all pigs were slaughtered, and colonic samples were collected. Results showed that dulcitol supplementation boosted intestinal barrier function in LPS-challenged piglets by enhancing intestinal morphology and integrity, and increasing the gene expression of zonula occludens-1, claudin-1, and occludin in the colonic mucosa (P <0.05). Metabolomics showed DUL supplementation mainly increased (P <0.05) the metabolites related to steroid and vitamin metabolism (Cholesterol and Vitamin C). Proteomics showed that dulcitol supplementation altered the protein expression involved in maintaining barrier integrity (FN1, CADM1, and PARD3), inhibiting inflammatory response (SLP1, SFN, and IRF3), and apoptosis (including FAS, ING1, BTK, MTHFR, NOX, and P53BP2) in LPS-challenged piglets (P <0.05). Additionally, dulcitol addition also suppressed the TLR4/NF-κB signaling pathway and apoptosis in mRNA and protein levels. Dulcitol increased the abundance of short-chain fatty acid-producing bacteria (Lactobacillus, Blautia, and Faecalibacterium) at the genus level, but decreased the relative abundance of Proteobacteria at the phylum level and Pseudomonas and Delftia at the genus level in piglets (P<.05). In conclusion, these results suggested that the addition of dulcitol alleviated LPS-induced intestinal barrier injury in piglets, probably by maintaining its integrity, inhibiting the TLR4/NF-κB signaling pathways and apoptosis, and modulating the gut microbiota. Therefore, dulcitol can be considered a potential dietary additive for improving intestinal health in pig models.


Assuntos
Suplementos Nutricionais , Mucosa Intestinal , Lipopolissacarídeos , Animais , Lipopolissacarídeos/toxicidade , Suínos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Enteropatias/induzido quimicamente , Enteropatias/prevenção & controle , Enteropatias/metabolismo , Intestinos/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Apoptose/efeitos dos fármacos , Masculino
5.
Life Sci ; 351: 122792, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-38857657

RESUMO

AIMS: Drug-induced enteropathy is often associated with the therapeutic use of certain glucuronidated drugs. One such drug is mycophenolic acid (MPA), a well-established immunosuppressant of which gastrointestinal adverse effects are a major concern. The role of bacterial ß-glucuronidase (ß-G) from the gut microbiota in MPA-induced enteropathy has recently been discovered. Bacterial ß-G hydrolyzes MPAG, the glucuronide metabolite of MPA excreted in the bile, leading to the digestive accumulation of MPA that would favor in turn these adverse events. We therefore hypothesized that taming bacterial ß-G activity might reduce MPA digestive exposure and prevent its toxicity. MAIN METHODS: By using a multiscale approach, we evaluated the effect of increasing concentrations of MPA on intestinal epithelial cells (Caco-2 cell line) viability, proliferation, and migration. Then, we investigated the inhibitory properties of amoxapine, a previously described bacterial ß-G inhibitor, by using molecular dynamics simulations, and evaluated its efficiency in blocking MPAG hydrolysis in an Escherichia coli-based ß-G activity assay. The pharmacological effect of amoxapine was evaluated in a mouse model. KEY FINDINGS: We observed that MPA impairs intestinal epithelial cell homeostasis. Amoxapine efficiently blocks the hydrolysis of MPAG to MPA and significantly reduces digestive exposure to MPA in mice. As a result, administration of amoxapine in MPA-treated mice significantly attenuated gastrointestinal lesions. SIGNIFICANCE: Collectively, these results suggest that the digestive accumulation of MPA is involved in the pathophysiology of MPA-gastrointestinal adverse effects. This study provides a proof-of-concept of the therapeutic potential of bacterial ß-G inhibitors in glucuronidated drug-induced enteropathy.


Assuntos
Biotransformação , Microbioma Gastrointestinal , Glucuronidase , Glucuronídeos , Ácido Micofenólico , Ácido Micofenólico/metabolismo , Ácido Micofenólico/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Glucuronidase/metabolismo , Glucuronidase/antagonistas & inibidores , Humanos , Animais , Camundongos , Glucuronídeos/metabolismo , Células CACO-2 , Masculino , Imunossupressores/farmacologia , Imunossupressores/toxicidade , Imunossupressores/metabolismo , Enteropatias/induzido quimicamente , Enteropatias/tratamento farmacológico , Enteropatias/metabolismo , Enteropatias/microbiologia , Proliferação de Células/efeitos dos fármacos , Glicoproteínas
6.
Poult Sci ; 103(8): 103964, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38936217

RESUMO

This study aimed to explore the impact of dietary Bacillus subtilis fmbj (BS) supplementation on acute intestinal dysfunction induced by lipopolysaccharide (LPS) in broilers. One hundred and eighty 1-day-old male Arbor Acres broilers were randomly divided into three treatment groups, each comprising ten replicates of 6 birds. On d 20, LPS-challenged (LPS group and LPS-BS group) and LPS-unchallenged (CON group) broilers received intraperitoneal injections of 1 mg/kg body weight LPS solution and an equivalent volume of sterile saline, respectively. Compared to the CON group, LPS disrupted (P < 0.05) the morphology of the small intestine (jejunum or ileum), exacerbated (P < 0.05) serum, small intestinal, and small intestinal mitochondrial antioxidant capacity, induced (P < 0.05) small intestinal oxidative damage, and altered (P < 0.05) the expression of genes and proteins related to antioxidants, cell adhesion, and mitochondrial function in the jejunum. The LPS-BS group exhibited a tendency towards improvement in small intestinal morphology, serum, small intestinal, and small intestinal mitochondrial antioxidant capacity, small intestinal oxidative damage, and the expression of genes and proteins related to antioxidants, cell adhesion, and mitochondrial function in the jejunum when compared to the LPS group. In conclusion, BS supplementation may confer protection against LPS-induced acute intestinal dysfunction in broilers by enhancing the activation of SIRT1/PGC1α, suggesting its potential as a valuable additive for the poultry industry.


Assuntos
Ração Animal , Bacillus subtilis , Galinhas , Dieta , Lipopolissacarídeos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Doenças das Aves Domésticas , Probióticos , Sirtuína 1 , Animais , Masculino , Bacillus subtilis/química , Doenças das Aves Domésticas/induzido quimicamente , Probióticos/administração & dosagem , Probióticos/farmacologia , Sirtuína 1/metabolismo , Sirtuína 1/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Ração Animal/análise , Dieta/veterinária , Enteropatias/veterinária , Enteropatias/induzido quimicamente , Distribuição Aleatória , Suplementos Nutricionais/análise , Proteínas Aviárias/metabolismo , Proteínas Aviárias/genética
9.
J Gastroenterol Hepatol ; 39(6): 1123-1133, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38576269

RESUMO

BACKGROUND AND AIM: Nonsteroidal anti-inflammatory drugs (NSAIDs) damage the small intestine via neutrophil infiltration driven by the mucosal invasion of enterobacteria. The antimicrobial function of neutrophils is partially dependent on neutrophil extracellular traps (NETs). Excessive NET formation has been associated with several inflammatory diseases. Here, we aimed to investigate the role of NETs in NSAID-induced small intestinal damage using human samples and an experimental mouse model. METHODS: Human small intestine specimens were obtained from NSAID users during double-balloon enteroscopy. Wild-type, protein arginine deiminase 4 (PAD4) knockout, and antibiotic-treated mice were administered indomethacin to induce small intestinal injury. The expression of NET-associated proteins, including PAD4, citrullinated histone H3 (CitH3), cell-free DNA, and myeloperoxidase (MPO), was evaluated. RESULTS: The double-positive stained area with CitH3 and MPO, which is specific for neutrophil-derived extracellular traps, was significantly high in the injured small intestinal mucosa of NSAID users. In a mouse model, small intestinal damage developed at 6 h after indomethacin administration, accompanied by increased mRNA levels of interleukin-1ß and keratinocyte chemoattractant and elevated NET-associated protein levels of PAD4, CitH3, and MPO in small intestine and serum levels of cell-free DNA. Both genetic deletion and pharmacological inhibition of PAD4 attenuated this damage by reducing the mRNA expression of inflammatory cytokines and NET-associated proteins. Furthermore, mice pretreated with antibiotics showed resistance to indomethacin-induced small intestinal damage, with less NET formation. CONCLUSION: These results suggest that NETs aggravate NSAID-induced small intestinal injury. Therefore, NET inhibition could be a potential treatment for NSAID-induced small intestinal injury.


Assuntos
Anti-Inflamatórios não Esteroides , Modelos Animais de Doenças , Armadilhas Extracelulares , Indometacina , Intestino Delgado , Peroxidase , Proteína-Arginina Desiminase do Tipo 4 , Animais , Armadilhas Extracelulares/metabolismo , Anti-Inflamatórios não Esteroides/efeitos adversos , Intestino Delgado/patologia , Intestino Delgado/metabolismo , Proteína-Arginina Desiminase do Tipo 4/metabolismo , Humanos , Indometacina/efeitos adversos , Peroxidase/metabolismo , Masculino , Neutrófilos/metabolismo , Histonas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Enteropatias/induzido quimicamente , Enteropatias/patologia , Interleucina-1beta/metabolismo , Mucosa Intestinal/patologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Camundongos Knockout , Feminino , Ácidos Nucleicos Livres/sangue , Citrulinação
10.
Poult Sci ; 103(5): 103586, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38442474

RESUMO

Intestinal inflammation is a primary contributor to poor growth performance during poultry production. Chlorogenic acid (CGA) is a natural phenolic acid that exhibits superior anti-inflammatory activity and improved intestinal health. To investigate the protective effects and molecular mechanisms of CGA during intestinal inflammation in lipopolysaccharide (LPS)-challenged broilers, we randomly divided 288 one-day-old male Cobb broilers into 4 groups: a control group fed a basal diet (CON group), a basal diet + LPS group (LPS group), and 2 basal diet groups fed 500 or 750 mg/kg CGA + LPS (CGA_500 or CGA_750 groups). Broilers were injected with LPS or saline at 15, 17, 19, and 21 d old. Chlorogenic acid supplementation improved the growth performance of LPS-challenged broilers by increasing average daily gain (ADG) and reducing feed/gain (F/G) ratios (P < 0.05). CGA also improved intestinal barrier function in LPS-challenged boilers by enhancing jejunum morphology and integrity, decreasing intestinal permeability, and increasing occludin 3, zonula occludens-1, and mucin 2 expression (P < 0.05). CGA supplementation also improved systemic and jejunum antioxidant capacity by significantly enhancing glutathione peroxidase (GSH-Px), superoxide dismutase (SOD), and catalase (CAT) activities (P < 0.05), and reducing malonaldehyde (MDA) and protein carbonyl (PCO) levels (P < 0.05). Chlorogenic acid supplementation reduced systemic and jejunum pro-inflammatory cytokines (interleukin (IL)-1ß, IL-6, and IL-12) and increased anti-inflammatory cytokines (IL-10) in LPS-challenged broilers (P < 0.05) by inhibiting the toll like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) signaling pathway (P < 0.05). In addition, the protective effects of CGA toward intestinal inflammation and apoptosis appeared to be correlated with inhibited endoplasmic reticulum (ER) stress (P < 0.05). In summary, CGA supplementation improved intestinal morphology and integrity by inhibiting TLR4/NF-κB and ER stress pathways, which potentially reduced oxidative stress and inflammation, and ultimately improved the growth performance of LPS-challenged broilers.


Assuntos
Galinhas , Ácido Clorogênico , Suplementos Nutricionais , Estresse do Retículo Endoplasmático , Lipopolissacarídeos , NF-kappa B , Doenças das Aves Domésticas , Animais , Ácido Clorogênico/administração & dosagem , Ácido Clorogênico/farmacologia , Lipopolissacarídeos/farmacologia , Masculino , NF-kappa B/metabolismo , Doenças das Aves Domésticas/induzido quimicamente , Doenças das Aves Domésticas/tratamento farmacológico , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Suplementos Nutricionais/análise , Dieta/veterinária , Inflamação/veterinária , Inflamação/tratamento farmacológico , Inflamação/induzido quimicamente , Distribuição Aleatória , Ração Animal/análise , Intestinos/efeitos dos fármacos , Intestinos/patologia , Enteropatias/veterinária , Enteropatias/induzido quimicamente , Enteropatias/tratamento farmacológico , Enteropatias/prevenção & controle , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/administração & dosagem
11.
In Vivo ; 38(2): 647-651, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38418161

RESUMO

BACKGROUND/AIM: Non-steroidal anti-inflammatory drugs (NSAIDs), the most widely used pharmaceuticals, induce various adverse effects, including gastrointestinal injuries, such as ulcers and bleeding. Animal models of NSAID-induced small intestinal injury (NSI) have been extensively employed for the development of preventive and therapeutic agents. However, some experimental variations related to feeding times have been observed following NSI induction. This study aimed to investigate the impact of feeding time on an NSI mouse model. MATERIALS AND METHODS: The mice were divided into eight groups: normal, sham, and model groups (with feeding times of 2 h, 6 h, 10 h, 14 h, 18 h, and 22 h; n=10 in each group). The mice were fasted for 18 h before the injection of indomethacin (15 mg/kg, subcutaneously), except for the normal group. Food supply was halted at specific time points (2 h, 6 h, 10 h, 14 h, 18 h, and 22 h); however, the normal and sham groups were continuously fed throughout the experiment. The length of the small intestine was measured, and histological analysis was performed 24 h after induction. RESULTS: Up to 14 h after induction, NSI, indicated by small intestine shortening, remained consistent, with a reduction in length of approximately 10-20%. However, feeding for more than 14 h significantly exacerbated NSI, both anatomically and histologically. CONCLUSION: The ulcerative changes observed in the small intestine 14 h after indomethacin injection may be closely associated with the influence of food on NSI.


Assuntos
Enteropatias , Camundongos , Animais , Enteropatias/induzido quimicamente , Enteropatias/patologia , Enteropatias/prevenção & controle , Anti-Inflamatórios não Esteroides/efeitos adversos , Indometacina/efeitos adversos , Intestino Delgado/patologia , Modelos Animais de Doenças , Úlcera/patologia
12.
Acta Pharmacol Sin ; 45(2): 339-353, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37816857

RESUMO

Lysophosphatidic acid (LPA) is a bioactive phospholipid mediator that has been found to ameliorate nonsteroidal anti-inflammatory drug (NSAID)-induced gastric injury by acting on lysophosphatidic acid type 2 receptor (LPAR2). In this study, we investigated whether LPAR2 signaling was implicated in the development of NSAID-induced small intestinal injury (enteropathy), another major complication of NSAID use. Wild-type (WT) and Lpar2 deficient (Lpar2-/-) mice were treated with a single, large dose (20 or 30 mg/kg, i.g.) of indomethacin (IND). The mice were euthanized at 6 or 24 h after IND treatment. We showed that IND-induced mucosal enteropathy and neutrophil recruitment occurred much earlier (at 6 h after IND treatment) in Lpar2-/- mice compared to WT mice, but the tissue levels of inflammatory mediators (IL-1ß, TNF-α, inducible COX-2, CAMP) remained at much lower levels. Administration of a selective LPAR2 agonist DBIBB (1, 10 mg/kg, i.g., twice at 24 h and 30 min before IND treatment) dose-dependently reduced mucosal injury and neutrophil activation in enteropathy, but it also enhanced IND-induced elevation of several proinflammatory chemokines and cytokines. By assessing caspase-3 activation, we found significantly increased intestinal apoptosis in IND-treated Lpar2-/- mice, but it was attenuated after DBIBB administration, especially in non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice. Finally, we showed that IND treatment reduced the plasma activity and expression of autotaxin (ATX), the main LPA-producing enzyme, and also reduced the intestinal expression of Lpar2 mRNA, which preceded the development of mucosal damage. We conclude that LPAR2 has a dual role in NSAID enteropathy, as it contributes to the maintenance of mucosal integrity after NSAID exposure, but also orchestrates the inflammatory responses associated with ulceration. Our study suggests that IND-induced inhibition of the ATX-LPAR2 axis is an early event in the pathogenesis of enteropathy.


Assuntos
Diabetes Mellitus Tipo 2 , Enteropatias , Lisofosfolipídeos , Camundongos , Animais , Receptores de Ácidos Lisofosfatídicos/genética , Receptores de Ácidos Lisofosfatídicos/metabolismo , Camundongos Endogâmicos NOD , Camundongos SCID , Anti-Inflamatórios não Esteroides , Indometacina/efeitos adversos , Enteropatias/induzido quimicamente
13.
BMC Gastroenterol ; 23(1): 449, 2023 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-38114956

RESUMO

BACKGROUND: Intestinal Behçet's disease (BD) is characterized by typical gastrointestinal ulcers in patients with BD followed by complications such as bleeding, perforation and fistula. Biologic agents are currently under active investigation to delay the disease course. Various data regarding infliximab are available, but there is relatively lack of data regarding adalimumab. METHODS: This was a multicenter, real-world prospective observational study to evaluate the effectiveness and safety of adalimumab in intestinal BD. The primary endpoint was disease activity at each follow up, including disease activity index for intestinal Behçet's disease (DAIBD), serum C-reactive protein (CRP) level, and endoscopic findings. The secondary endpoint was the incidence of adverse drug reactions (ADRs). RESULTS: A total of 58 patients were enrolled and 8 of them were excluded. Adverse events were reported in 72.0% of patients with 122 events. ADRs were reported in 24.0% with 28 events. For adverse events, arthralgia was most commonly reported (13.1%: 16/122) and only one experienced critical adverse event (0.82%, 1/122: death due to stroke). On multivariable regression analysis, a longer disease duration was significantly associated with decreased ADRs [Odds ratio 0.976 (0.953-0.999, 95% CI); p = 0.042]. Clinical response rates as assessed by DAIBD were 90.9% at Week 12 and 89.7% at Week 56, respectively. The mean serum CRP level at baseline was significantly decreased after 12 weeks (3.91 ± 4.93 to 1.26 ± 2.03 mg/dL; p = 0.0002). CONCLUSION: Adalimumab was found to be safe and effective in Korean patients with intestinal BD. A longer disease duration was significantly associated with decreased ADRs.


Assuntos
Síndrome de Behçet , Enteropatias , Humanos , Adalimumab/efeitos adversos , Síndrome de Behçet/complicações , Síndrome de Behçet/tratamento farmacológico , Intestinos , Infliximab , Enteropatias/tratamento farmacológico , Enteropatias/induzido quimicamente
14.
Benef Microbes ; 14(3): 239-253, 2023 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-37646075

RESUMO

Nonsteroidal anti-inflammatory drugs (NSAIDs) induce a broad spectrum of gastro-intestinal adverse effects, including ulceration and bleeding. The pathophysiology of NSAID enteropathy is complex and incompletely understood, but some evidence showed that NSAIDs impair the intestinal barrier and cause a gut dysbiosis. Identifying new treatments aiming to reverse or attenuate NSAID-induced adverse effects would have a significant impact on a high number of patients. The aim of this work is to assess the effects of the probiotic yeast Saccharomyces boulardii CNCM I-745 (Sb) on a model of NSAID-induced enteropathy. Four groups of mice were tested: Control, Indomethacin, Sb, and Sb + Indomethacin. A clinical score was evaluated throughout the experiment. Faecal calprotectin, microbiota and haemoglobin analyses were performed. At the end of the treatments, the small intestine, colon, and caecum lengths, and intestinal permeability were measured. Sections of ileum and jejunum were observed to assess a histological score and ileal cytokines were measured by immunoassay. Indomethacin-treated animals showed an increase in their clinical scores, reflecting a worsening of their general state. Mice co-treated with Sb and indomethacin displayed an improvement of their clinical score in comparison with mice treated with indomethacin alone. Sb prevented the indomethacin-induced shortening of the small intestine and caecum, and significantly attenuated the severity of intestinal lesions. Sb also prevented the increase in faecal calprotectin, reduced faecal haemoglobin, and prevented the increase of intestinal permeability in mice treated with indomethacin. Sb also counteracted the increase of faecal bacteria associated with the pathogenesis of NSAID-enteropathy. In conclusion, our results show a protective effect of Sb in a model of indomethacin-induced enteropathy. Sb improved the intestinal barrier function and exerted a positive action on gut microbiota composition.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Enteropatias , Probióticos , Saccharomyces boulardii , Humanos , Animais , Camundongos , Anti-Inflamatórios não Esteroides/efeitos adversos , Indometacina/toxicidade , Saccharomyces cerevisiae , Enteropatias/induzido quimicamente , Enteropatias/prevenção & controle , Modelos Teóricos , Hemoglobinas , Complexo Antígeno L1 Leucocitário
15.
Food Funct ; 14(16): 7705-7717, 2023 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-37547959

RESUMO

During weaning, piglets are susceptible to intestinal injuries caused by a range of infections, which result in serious economic losses for pig producers. Caffeic acid (CA) is a plant-derived phenolic acid that exhibits potential as a dietary supplement for enhancing intestinal health. There is, however, limited information available about the potential benefits of CA supplementation on intestinal injury and growth performance in piglets. A 28-day study was conducted to examine the effectiveness of CA supplementation in protecting against intestinal injury induced by intraperitoneal injection of Escherichia coli lipopolysaccharide (LPS) in piglets. Twenty-four piglets (7.43 ± 0.79 kg body weight; Duroc × Landrace × Large White; barrows) were randomly divided into 4 groups: the control group, the LPS group, the LPS + CA group, and the CA group. Piglets were administered with LPS or saline on d21 and d28 of the experiment. Supplementation with CA improved intestinal barrier function in LPS-challenged piglets by enhancing intestinal morphology and integrity, as well as increasing the expression of Claudin-1 and ZO-1. Meanwhile, CA supplementation improved the systemic and colonic inflammation responses, oxidative stress, and apoptosis induced by LPS. CA supplementation improved the alpha diversity and structure of the intestinal microbiota by increasing the abundance of beneficial microbiota. Additionally, it was found that it improves metabolic disorders of colonic bile acids (BAs) and short-chain fatty acids (SCFAs) in LPS-challenged piglets, including an increase in primary BAs and isovalerate. In conclusion, CA supplementation could enhance intestinal integrity and barrier function by modifying intestinal microbiota and its metabolites, which could lead to a reduction in inflammatory responses and oxidative stress and ultimately enhanced growth performance in piglets.


Assuntos
Microbioma Gastrointestinal , Enteropatias , Suínos , Animais , Lipopolissacarídeos/efeitos adversos , Suplementos Nutricionais/análise , Intestinos , Enteropatias/tratamento farmacológico , Enteropatias/veterinária , Enteropatias/induzido quimicamente , Desmame
16.
Br J Pharmacol ; 180(24): 3215-3233, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37519261

RESUMO

BACKGROUND AND PURPOSE: Nonsteroidal anti-inflammatory drugs (NSAIDs) can be associated with severe adverse digestive effects. This study examined the protective effects of the probiotic Saccharomyces boulardii CNCM I-745 in a rat model of diclofenac-induced enteropathy. EXPERIMENTAL APPROACH: Enteropathy was induced in 40-week-old male rats by intragastric diclofenac (4 mg·kg-1 BID for 14 days). S. boulardii CNCM I-745 (3 g·kg-1 BID by oral gavage) was administered starting 14 days before (preventive protocol) or along with (curative protocol) diclofenac administration. Ileal damage, inflammation, barrier integrity, gut microbiota composition and toll-like receptors (TLRs)-nuclear factor κB (NF-κB) pathway were evaluated. KEY RESULTS: Diclofenac elicited intestinal damage, along with increments of myeloperoxidase, malondialdehyde, tumour necrosis factor and interleukin-1ß, overexpression of TLR2/4, myeloid differentiation primary response 88 (Myd88) and NF-κB p65, increased faecal calprotectin and butyrate levels, and decreased blood haemoglobin levels, occludin and butyrate transporter monocarboxylate transporter 1 (MCT1) expression. In addition, diclofenac provoked a shift of bacterial taxa in both faecal and ileal samples. Treatment with S. boulardii CNCM I-745, in both preventive and curative protocols, counteracted the majority of these deleterious changes. Only preventive administration of the probiotic counteracted NSAID-induced decreased expression of MCT1 and increase in faecal butyrate levels. Occludin expression, after probiotic treatment, did not significantly change. CONCLUSIONS AND IMPLICATIONS: Treatment with S. boulardii CNCM I-745 prevents diclofenac-induced enteropathy through anti-inflammatory and antioxidant activities. Such effects are likely to be related to increased tissue butyrate bioavailability, through an improvement of butyrate uptake by the enteric mucosa.


Assuntos
Enteropatias , Saccharomyces boulardii , Masculino , Ratos , Animais , Saccharomyces boulardii/fisiologia , Diclofenaco , NF-kappa B , Ocludina , Enteropatias/induzido quimicamente , Enteropatias/prevenção & controle , Anti-Inflamatórios não Esteroides , Butiratos
17.
Medicine (Baltimore) ; 102(25): e34118, 2023 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-37352037

RESUMO

RATIONALE: Tocilizumab, a humanized anti-interleukin-6 (IL-6) receptor monoclonal antibody, is used for the treatment of adult-onset Still disease (AOSD). Despite its efficacy in many clinical situations, concerns have been raised regarding intestinal mucosal injury in patients receiving tocilizumab. PATIENT CONCERNS: A 64-year-old woman with a history of AOSD was admitted to our hospital with hematochezia. She had AOSD for 15 years and underwent treatment with biweekly tocilizumab 9 months prior to admission. Colonoscopy revealed a large punched-out ulcer in the terminal ileum. On pathological evaluation, nonspecific enteritis with lymphocytes and eosinophils were seen. Based on the location and shape of the lesion, we suspected intestinal Behçet's disease. However, the ulcer reduced in size over time by discontinuation of tocilizumab without additional drug treatment, indicating that it was a drug-induced ulcer. DIAGNOSIS: The patient was diagnosed with tocilizumab-induced small intestinal ulcer. INTERVENTIONS: The patient treated with the discontinuation of tocilizumab. OUTCOMES: The discontinuation of tocilizumab resulted in ulcer scarring. There was no recurrence of hematochezia. LESSONS: Tocilizumab can cause deep ulcerative lesions in the terminal ileum, which may resemble intestinal Behçet's disease. It is important to continuously monitor abdominal symptoms during tocilizumab therapy and aggressively perform colonoscopy when hematochezia or abdominal pain is observed.


Assuntos
Síndrome de Behçet , Enteropatias , Adulto , Feminino , Humanos , Pessoa de Meia-Idade , Síndrome de Behçet/tratamento farmacológico , Úlcera/induzido quimicamente , Úlcera/diagnóstico , Úlcera/tratamento farmacológico , Enteropatias/induzido quimicamente , Enteropatias/diagnóstico , Enteropatias/tratamento farmacológico , Anticorpos Monoclonais/uso terapêutico , Íleo/patologia , Hemorragia Gastrointestinal/tratamento farmacológico
18.
Int Immunopharmacol ; 119: 110205, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37104917

RESUMO

Irinotecan (CPT-11) is a topoisomerase I inhibitor that was approved for cancer treatment in 1994. To date, this natural product derivative remains the world's leading antitumor drug. However, the clinical application of irinotecan is limited due to its side effects, the most troubling of which is intestinal toxicity. In addition, irinotecan has certain toxicity to cells and even causes cellular senescence. Committed to developing alternatives to prevent these adverse reactions, we evaluated the activity of artesunate, which has never been tested in this regard despite its biological potential. Irinotecan accelerated the process of aging in vivo and in vitro, and we found that this was mainly caused by activating mTOR signaling targets. Artesunate inhibited the activity of mTOR, thereby alleviating the aging process. Our study found that artesunate treatment improved irinotecan-induced intestinal inflammation by reducing the levels of TNF-α, IL1, and IL6; reducing inflammatory infiltration of the colonic ileum in mice; and preventing irinotecan-induced intestinal damage by reducing weight loss and improving intestinal length. In addition, in mouse xenograft tumor models, artesunate and irinotecan significantly inhibited tumor growth in mice.


Assuntos
Antineoplásicos , Artesunato , Enteropatias , Irinotecano , Inibidores da Topoisomerase I , Artesunato/uso terapêutico , Humanos , Animais , Camundongos , Antineoplásicos/uso terapêutico , Irinotecano/efeitos adversos , Enteropatias/induzido quimicamente , Enteropatias/tratamento farmacológico , Senescência Celular , Inibidores da Topoisomerase I/efeitos adversos
19.
Clin Transl Gastroenterol ; 14(6): e00588, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37019683

RESUMO

INTRODUCTION: Conflicting results exist on the association between proton-pump inhibitor (PPI) and nonsteroidal anti-inflammatory drug (NSAID)-related small-bowel damage. The aim of this study was to determine whether PPIs increased the risk of NSAID-related small-bowel damage by meta-analysis. METHODS: A systematic electronic search in PubMed, Embase, and Web of Science was conducted from the time the database was created until March 31, 2022, for studies reporting associations between PPI use and outcomes, including the endoscopy-verified prevalence of small-bowel injury, mean number of small-bowel injuries per patient, change in hemoglobin level, and risk of small-bowel bleeding in subjects taking NSAIDs. Meta-analytical calculations for odds ratio (OR) and mean difference (MD) were performed with the random-effects model and interpreted with 95% confidence intervals (CIs). RESULTS: Fourteen studies comprising 1996 subjects were included. Pooled analysis demonstrated that concomitant use of PPIs significantly increased the prevalence and number of endoscopy-verified small-bowel injuries (prevalence: OR = 3.00; 95% CI: 1.74-5.16; number: MD = 2.30; 95% CI: 0.61-3.99) and decreased hemoglobin levels (MD = -0.50 g/dL; 95% CI: 0.88 to -0.12) in NSAID users but did not change the risk of small-bowel bleeding (OR = 1.24; 95% CI: 0.80-1.92). Subgroup analysis demonstrated that PPIs significantly increased the prevalence of small-bowel injury in subjects taking nonselective NSAIDs (OR = 7.05; 95% CI: 4.70-10.59, 4 studies, I 2 = 0) and COX-2 inhibitors (OR = 4.00; 95% CI: 1.18-13.60, 1 study, no calculated I 2 ) when compared with COX-2 inhibitors alone. DISCUSSION: PPIs increased the risk of NSAID-related small-bowel damage, and the clinical significance of higher prevalence of small-bowel injuries should be studied in the future.


Assuntos
Enteropatias , Inibidores da Bomba de Prótons , Humanos , Inibidores da Bomba de Prótons/efeitos adversos , Inibidores de Ciclo-Oxigenase 2 , Anti-Inflamatórios não Esteroides/efeitos adversos , Enteropatias/induzido quimicamente , Enteropatias/epidemiologia , Hemorragia Gastrointestinal/induzido quimicamente , Hemorragia Gastrointestinal/epidemiologia , Hemoglobinas
20.
Pharmacology ; 108(3): 286-300, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37023725

RESUMO

INTRODUCTION: Nonsteroidal anti-inflammatory drug (NSAID)-induced small intestinal damage is a serious and escalating clinical problem without effective treatment. Lafutidine (LAF) is a novel histamine H2 receptor antagonist with a mucosal protective action. This study aimed to investigate the protective effect of LAF on indomethacin (IND)-induced enteropathy in rats. METHODS: Rats were treated with LAF for 10 days with concomitant IND treatment on the final 5 days. Changes in metabolism and hematological and biochemical parameters were measured, and intestinal damage was blindly scored. Intestinal mucosal tissue and luminal contents were collected for transcriptome and microbiota sequencing. Intestinal inflammation and barrier function were also evaluated. RESULTS: LAF treatment prevented anorexia and weight loss in rats and ameliorated reductions in hemoglobin, hematocrit, total protein, and albumin levels. LAF reduced the severity of IND-induced intestinal damage including macroscopic and histopathological damage score. Transcriptome sequencing results indicated that LAF might have positive effects on intestinal inflammation and the intestinal mucosal barrier. Further research revealed that LAF decreased neutrophil infiltration, and IL-1ß and TNF-α expression in intestinal tissue. Besides, the treatment increased mucus secretion, MUC2, Occludin, and ZO-1 expression, and decreased serum D-lactate levels. LAF treatment also ameliorates microbial dysbiosis in small intestine induced by IND and increased the abundance of Lactobacillus acidophilus. CONCLUSION: LAF may protect against NSAID enteropathy via enhancing the intestinal mucosal barrier, inhibiting inflammation, and regulating microbiota.


Assuntos
Enteropatias , Microbiota , Ratos , Animais , Indometacina/toxicidade , Intestino Delgado , Anti-Inflamatórios não Esteroides/farmacologia , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Mucosa Intestinal , Enteropatias/induzido quimicamente
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...