1
|
Liu P, Yang J, Jin M, Hu P, Zhu Y, Tang Y, Chen Y, Xu X, He H. Alterations in the gut microbiome and metabolism profiles reveal the possible molecular mechanism of renal injury induced by hyperuricemia in a mouse model of renal insufficiency. Ren Fail 2024; 46:2387429. [PMID: 39132829 PMCID: PMC11321104 DOI: 10.1080/0886022x.2024.2387429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/12/2023] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/13/2024] Open
Abstract
Objectives: To investigate the role of the intestinal flora and metabolites in the development of hyperuricemic renal injury in chronic kidney disease (CKD).Methods: Unilaterally nephrectomized mice were fed with adenine and potassium oxonate for 9 weeks. HE staining combined with plasma biochemical indicators was used to evaluate renal pathological and functional changes. We conducted 16S rRNA sequencing and untargeted metabolomics on feces and plasma samples to reveale changes in intestinal microbiota and metabolites.Result: Our analysis revealed significant differences in 15 bacterial genera, with 7 being upregulated and 8 being downregulated. Furthermore, metabolomic analysis revealed changes in the distribution of amino acid and biotin metabolites in basic metabolic pathways in both feces and serum. Specifically, differentially abundant metabolites in feces were associated primarily with histidine metabolism; the biosynthesis of phenylalanine, tyrosine, and tryptophan; and tyrosine metabolism. In plasma, the differentially abundant metabolites were involved in multiple metabolic pathways, including aminoacyl-tRNA biosynthesis; glycine, serine, and threonine amino acid metabolism; valine, leucine, and isoleucine biosynthesis; tyrosine biosynthesis and metabolism; biotin metabolism; and taurine and hypotaurine metabolism. Furthermore, correlation analysis revealed that Akkermansia, UCG-005, Lachnospiraceae_NK4A136_group, Lactococcus, and Butymonas were associated with various differentially abundant metabolites as well as renal function, oxidative stress, and mitophagy. The changes in the intestinal flora observed in hyperuricemia may lead to imbalances in amino acid and biotin metabolism in both the intestine and host, ultimately affecting oxidative stress and mitophagy in mice and accelerating the progression of CKD.Conclusion: Our findings provide insights into a potential pathogenic mechanism by which hyperuricemia exacerbates renal injury in mice with renal insufficiency. Understanding these pathways may offer new therapeutic strategies for managing hyperuricemic renal injury in CKD patients.
Collapse
Affiliation(s)
- Ping Liu
- Division of Nephrology, Minhang Hospital, Fudan University, Shanghai, China
| | - Jianli Yang
- East China University of Science and Technology, Shanghai, China
| | - Meiping Jin
- Division of Nephrology, Minhang Hospital, Fudan University, Shanghai, China
| | - Ping Hu
- Division of Nephrology, Minhang Hospital, Fudan University, Shanghai, China
| | - Yifan Zhu
- Division of Nephrology, Minhang Hospital, Fudan University, Shanghai, China
| | - Yuyan Tang
- Division of Nephrology, Minhang Hospital, Fudan University, Shanghai, China
| | - Yu Chen
- East China University of Science and Technology, Shanghai, China
| | - Xudong Xu
- Division of Nephrology, Minhang Hospital, Fudan University, Shanghai, China
| | - Haidong He
- Division of Nephrology, Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Stojiljković S, Gavrilović L, Pejić S, Pajović SB, Macura M, Nikolić D, Bubanj S, Stojiljković V. Effects of Endurance Training on Antioxidant and Hormonal Status in Peripheral Blood of Young Healthy Men. Life (Basel) 2024; 14:921. [PMID: 39202664 PMCID: PMC11355762 DOI: 10.3390/life14080921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/11/2024] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 09/03/2024] Open
Abstract
(1) Background: Physical activity may cause an imbalance in the major functions of the human body. This study aimed to investigate the effects of endurance running training on the parameters of the antioxidant defense system (SOD, CAT, GPx, GR, GSH), LPO (malondialdehyde, MDA), and stress hormones (A, NA) in young healthy, previously untrained men. (2) Methods: The training program was as follows: 8 weeks of running, three times per week; the duration of a single session was 30-70 min, the intensity was twice a week in the so-called extensive endurance zone, and once a week in the anaerobic threshold zone. Blood samples were collected from the subjects, before and after the running program. (3) Results: The training program resulted in a significant increase in maximal oxygen consumption (p < 0.001). The activities of SOD, GPx, and GR also increased significantly (p < 0.05, p < 0.01, and p < 0.05, respectively), while CAT activity and GSH and MDA concentrations remained unchanged. The concentration of A decreased (p < 0.05), while the NA concentration increased significantly (p < 0.05). SOD, GPx, GR, and NA positively correlated with VO2max (p < 0.05, p < 0.001, p < 0.01, p < 0.05, respectively), while a negative correlation was detected between A and VO2max (p < 0.05). (4) Conclusions: These results indicate that there is no persistent oxidative stress in response to the applied 8-week running program, probably due to exercise-induced protective alterations in the antioxidant defense system. Furthermore, adaptations occurred at the hormonal level, making the organism more ready for a new challenge.
Collapse
Affiliation(s)
- Stanimir Stojiljković
- Faculty of Sport and Physical Education, University of Belgrade, 11000 Belgrade, Serbia; (S.S.); (M.M.)
| | - Ljubica Gavrilović
- Department of Molecular Biology and Endocrinology, “Vinča” Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (L.G.); (S.P.); (S.B.P.)
| | - Snežana Pejić
- Department of Molecular Biology and Endocrinology, “Vinča” Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (L.G.); (S.P.); (S.B.P.)
| | - Snežana B. Pajović
- Department of Molecular Biology and Endocrinology, “Vinča” Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (L.G.); (S.P.); (S.B.P.)
| | - Marija Macura
- Faculty of Sport and Physical Education, University of Belgrade, 11000 Belgrade, Serbia; (S.S.); (M.M.)
| | - Dragan Nikolić
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Clinic for Endocrinology, Diabetes and Metabolic Diseases (Laboratory for Cells Culture), Clinical Centre of Serbia, 11000 Belgrade, Serbia
| | - Saša Bubanj
- Faculty of Sport and Physical Education, University of Niš, 18000 Niš, Serbia;
| | - Vesna Stojiljković
- Department of Molecular Biology and Endocrinology, “Vinča” Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (L.G.); (S.P.); (S.B.P.)
| |
Collapse
|
3
|
Liu X, Zeng X, Li X, Xin S, Zhang F, Liu F, Zeng Y, Wu J, Zou Y, Xiong X. Landscapes of gut bacterial and fecal metabolic signatures and their relationship in severe preeclampsia. J Transl Med 2024; 22:360. [PMID: 38632606 PMCID: PMC11022388 DOI: 10.1186/s12967-024-05143-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/05/2024] [Accepted: 03/28/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Preeclampsia is a pregnancy-specific disease leading to maternal and perinatal morbidity. Hypertension and inflammation are the main characteristics of preeclampsia. Many factors can lead to hypertension and inflammation, including gut microbiota which plays an important role in hypertension and inflammation in humans. However, alterations to the gut microbiome and fecal metabolome, and their relationships in severe preeclampsia are not well known. This study aims to identify biomarkers significantly associated with severe preeclampsia and provide a knowledge base for treatments regulating the gut microbiome. METHODS In this study, fecal samples were collected from individuals with severe preeclampsia and healthy controls for shotgun metagenomic sequencing to evaluate changes in gut microbiota composition. Quantitative polymerase chain reaction analysis was used to validate the reliability of our shotgun metagenomic sequencing results. Additionally, untargeted metabolomics analysis was performed to measure fecal metabolome concentrations. RESULTS We identified several Lactobacillaceae that were significantly enriched in the gut of healthy controls, including Limosilactobacillus fermentum, the key biomarker distinguishing severe preeclampsia from healthy controls. Limosilactobacillus fermentum was significantly associated with shifts in KEGG Orthology (KO) genes and KEGG pathways of the gut microbiome in severe preeclampsia, such as flagellar assembly. Untargeted fecal metabolome analysis found that severe preeclampsia had higher concentrations of Phenylpropanoate and Agmatine. Increased concentrations of Phenylpropanoate and Agmatine were associated with the abundance of Limosilactobacillus fermentum. Furthermore, all metabolites with higher abundances in healthy controls were enriched in the arginine and proline metabolism pathway. CONCLUSION Our research indicates that changes in metabolites, possibly due to the gut microbe Limosilactobacillus fermentum, can contribute to the development of severe preeclampsia. This study provides insights into the interaction between gut microbiome and fecal metabolites and offers a basis for improving severe preeclampsia by modulating the gut microbiome.
Collapse
Affiliation(s)
- Xianxian Liu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, 330006, China
| | - Xiaoming Zeng
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, 330006, China
| | - Xing Li
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Siming Xin
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, 330006, China
| | - Feng Zhang
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, 330006, China
| | - Faying Liu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, 330006, China
| | - Yang Zeng
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, 330006, China
| | - Jilin Wu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, 330006, China
| | - Yang Zou
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, 330006, China.
| | - Xinwei Xiong
- Institute of Biological Technology, Nanchang Normal University, Nanchang, Jiangxi, 330032, China.
| |
Collapse
|
4
|
Sestak SS, da Motta Lima FG, de Oliveira AP, Barateiro LGRP, Vieira-Frez FC, de Souza SRG, Guarnier FA, Perles JVCM, Zanoni JN. Effects of cancer-induced cachexia and administration of L-glutathione on the intestinal mucosa in rat. Amino Acids 2024; 56:30. [PMID: 38607556 PMCID: PMC11009745 DOI: 10.1007/s00726-024-03391-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/09/2024] [Accepted: 03/19/2024] [Indexed: 04/13/2024]
Abstract
Walker-256 tumor is an experimental model known to promote cachexia syndrome, oxidative stress, and systemic inflammation. This study evaluated the duodenal mucosa of rats with Walker-256 tumor administered with 1% L-glutathione, intending to evaluate the damage caused by cancer-associated cachexia in the gastrointestinal tract and the effects of antioxidant administration on mucosal protection. Twenty-four 55-day-old male Wistar rats were distributed into four groups: control (C); control administered with 1% L-glutathione (C-GSH); Walker-256 tumor (W) and Walker-256 tumor administered with 1% L-glutathione (W-GSH). After 14 days of treatment, the duodenum was harvested for morphometric analysis of the mucosa, proliferation, apoptosis, immunostaining of varicosities immunoreactive (IR) to vasoactive intestinal peptide (VIP) and 5-HT-IR cells, and quantification of mast cells and goblet cells. Walker-256 tumor-bearing rats showed cachexia syndrome, mucosal atrophy, reduced cell proliferation, reduced 5-HT-IR cells, and increased goblet cells and VIPergic varicosities, which were not reversed by L-glutathione. On the other hand, L-glutathione caused a reduction of cells in apoptosis and mast cell recruitment, demonstrating a partial recovery of the damage detected in the intestinal mucosa.
Collapse
Affiliation(s)
- Sabrina Silva Sestak
- Department of Physiology, Laboratory of Enteric Neural Plasticity, State University of Maringá, O33 Block, Colombo Avenue, 5790, Maringá, Paraná, CEP 87020-900, Brazil
| | - Fabiana Galvão da Motta Lima
- Department of Physiology, Laboratory of Enteric Neural Plasticity, State University of Maringá, O33 Block, Colombo Avenue, 5790, Maringá, Paraná, CEP 87020-900, Brazil
| | - Ana Paula de Oliveira
- Department of Physiology, Laboratory of Enteric Neural Plasticity, State University of Maringá, O33 Block, Colombo Avenue, 5790, Maringá, Paraná, CEP 87020-900, Brazil
| | | | | | | | | | | | - Jacqueline Nelisis Zanoni
- Department of Physiology, Laboratory of Enteric Neural Plasticity, State University of Maringá, O33 Block, Colombo Avenue, 5790, Maringá, Paraná, CEP 87020-900, Brazil.
- Department of Morphological Sciences, State University of Maringá, Maringá, Paraná, Brazil.
| |
Collapse
|
5
|
Lima FGDM, Silva MPAD, Sestak SS, Guarnier FA, de Oliveira AP, Kuller JV, Gulbransen BD, Perles JVCM, Zanoni JN. Cancer-induced morphological changes in enteric glial cells in the jejunum of Walker-256 tumor-bearing rats. Acta Histochem 2024; 126:152146. [PMID: 38422841 PMCID: PMC11039380 DOI: 10.1016/j.acthis.2024.152146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/26/2023] [Revised: 01/29/2024] [Accepted: 02/14/2024] [Indexed: 03/02/2024]
Abstract
Cancer-induced cachexia is associated with systemic inflammation and gastrointestinal dysfunction. How changes to cells of the enteric nervous system contribute to gut dysfunction in tumor development and cancer cachexia is unknown. Here, we tested the hypothesis that changes to enteric glia, a type of peripheral glia that surround enteric neurons and regulate gut homeostasis, are associated with tumor development and that supplementing with the antioxidant L-glutathione is protective against the changes induced. Immunohistochemistry for neurons, enteric glial cells and immune cells was performed in whole-mount preparations and frozen histological sections of the jejunum from 20 Wistar rats, distributed in 4 groups: control, tumor of Walker-256, control administered with 1 % L-glutathione, and tumor of Walker-256 administered with 1 % L-glutathione. Morphoquantitative analyses were made using Image-Pro® Plus 4.5 and ImageJ® 1.43° software. Tumor development significantly reduced neuronal and glial cell populations in the myenteric and submucosal plexuses and enlarged glial cell body area in the submucosal plexus. In contrast, tumors increased glia in the jejunal mucosa and this effect was accompanied by B-lymphocyte recruitment. GSH-supplemented diet was not sufficient to protect against changes to neurons and glia in the submucosal plexus but was partially protective in the myenteric plexus. L-glutathione had no effect on physiological parameters of cachexia but was sufficient to preserve enteric glial cell density in the myenteric plexus. These results suggest that changes to both enteric neurons and glia likely contribute to the gastrointestinal effects of tumor development and that oxidative stress contributes to these effects in the enteric nervous system.
Collapse
Affiliation(s)
| | | | - Sabrina Silva Sestak
- Laboratory of Enteric Neural Plasticity, State University of Maringá, Maringá, PR, Brazil
| | | | | | - João Victor Kuller
- Laboratory of Enteric Neural Plasticity, State University of Maringá, Maringá, PR, Brazil
| | | | | | | |
Collapse
|
6
|
Li J, Zhang J, Jin X, Li S, Du Y, Zeng Y, Wang J, Chen W. Identification and functional prediction of long non-coding RNAs related to oxidative stress in the jejunum of piglets. Anim Biosci 2024; 37:193-202. [PMID: 37641831 PMCID: PMC10766486 DOI: 10.5713/ab.23.0202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/26/2023] [Revised: 07/18/2023] [Accepted: 07/31/2023] [Indexed: 08/31/2023] Open
Abstract
OBJECTIVE Oxidative stress (OS) is a pathological process arising from the excessive production of free radicals in the body. It has the potential to alter animal gene expression and cause damage to the jejunum. However, there have been few reports of changes in the expression of long noncoding RNAs (lncRNAs) in the jejunum in piglets under OS. The purpose of this research was to examine how lncRNAs in piglet jejunum change under OS. METHODS The abdominal cavities of piglets were injected with diquat (DQ) to produce OS. Raw reads were downloaded from the SRA database. RNA-seq was utilized to study the expression of lncRNAs in piglets under OS. Additionally, six randomly selected lncRNAs were verified using quantitative real-time polymerase chain reaction (qRT‒PCR) to examine the mechanism of oxidative damage. RESULTS A total of 79 lncRNAs were differentially expressed (DE) in the treatment group compared to the negative control group. The target genes of DE lncRNAs were enriched in gene ontology (GO) terms and Kyoto encyclopedia of genes and genomes (KEGG) signaling pathways. Chemical carcinogenesis-reactive oxygen species, the Foxo signaling pathway, colorectal cancer, and the AMPK signaling pathway were all linked to OS. CONCLUSION Our results demonstrated that DQ-induced OS causes differential expression of lncRNAs, laying the groundwork for future research into the processes involved in the jejunum's response to OS.
Collapse
Affiliation(s)
- Jinbao Li
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Tai’an City, 271018,
China
| | - Jianmin Zhang
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Tai’an City, 271018,
China
| | - Xinlin Jin
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Tai’an City, 271018,
China
| | - Shiyin Li
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Tai’an City, 271018,
China
| | - Yingbin Du
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Tai’an City, 271018,
China
| | - Yongqing Zeng
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Tai’an City, 271018,
China
| | - Jin Wang
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Tai’an City, 271018,
China
| | - Wei Chen
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Tai’an City, 271018,
China
| |
Collapse
|
7
|
Zhu C, Nie X, Lu Q, Bai Y, Jiang Z. Roles and regulation of Aquaporin-3 in maintaining the gut health: an updated review. Front Physiol 2023; 14:1264570. [PMID: 38089478 PMCID: PMC10714013 DOI: 10.3389/fphys.2023.1264570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/21/2023] [Accepted: 11/15/2023] [Indexed: 01/05/2025] Open
Abstract
Aquaporin-3 (AQP3) is a predominant water channel protein expressed in the intestine, and plays important roles in the gut physiology and pathophysiology due to its permeability to water, glycerol and hydrogen peroxide. In this review, we systematically summarized the current understanding of the expression of AQP3 in the intestine of different species, and focused on the potential roles of AQP3 in water transport, different types of diarrhea and constipation, intestinal inflammation, intestinal barrier function, oxidative stress, and autophagy. These updated findings have supported that AQP3 may function as an important target in maintaining gut health of human and animals.
Collapse
Affiliation(s)
- Cui Zhu
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Xiaoyan Nie
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Qi Lu
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Yinshan Bai
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Zongyong Jiang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| |
Collapse
|
8
|
Abstract
The gastrointestinal (GI) barrier acts as the primary interface between humans and the external environment. It constantly faces the risk of inflammation and oxidative stress due to exposure to foreign substances and microorganisms. Thus, maintaining the structural and functional integrity of the GI barrier is crucial for overall well-being, as it helps prevent systemic inflammation and oxidative stress, which are major contributors to age-related diseases. A healthy gut relies on maintaining gut redox homeostasis, which involves several essential elements. Firstly, it requires establishing a baseline electrophilic tone and an electrophilic mucosal gradient. Secondly, the electrophilic system needs to have sufficient capacity to generate reactive oxygen species, enabling effective elimination of invading microorganisms and rapid restoration of the barrier integrity following breaches. These elements depend on physiological redox signaling mediated by electrophilic pathways such as NOX2 and the H2O2 pathway. Additionally, the nucleophilic arm of redox homeostasis should exhibit sufficient reactivity to restore the redox balance after an electrophilic surge. Factors contributing to the nucleophilic arm include the availability of reductive substrates and redox signaling mediated by the cytoprotective Keap1-Nrf2 pathway. Future research should focus on identifying preventive and therapeutic strategies that enhance the strength and responsiveness of GI redox homeostasis. These strategies aim to reduce the vulnerability of the gut to harmful stimuli and address the decline in reactivity often observed during the aging process. By strengthening GI redox homeostasis, we can potentially mitigate the risks associated with age-related gut dyshomeostasis and optimize overall health and longevity.
Collapse
Affiliation(s)
- Jan Homolak
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia.
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 12, 10 000, Zagreb, Croatia.
| |
Collapse
|
9
|
Liang H, Xie Y, Li M, Chen J, Zhou W, Xia R, Ding Q, Yao Y, Zhang Z, Yang Y, Ran C, Zhou Z. The effect of stabilized culture of Lactobacillus rhamnosus GCC-3 on gut and liver health, and anti-viral immunity of zebrafish. FISH & SHELLFISH IMMUNOLOGY 2023; 141:109074. [PMID: 37714442 DOI: 10.1016/j.fsi.2023.109074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 06/16/2023] [Revised: 07/31/2023] [Accepted: 09/11/2023] [Indexed: 09/17/2023]
Abstract
Probiotics are promising antibiotics alternatives to improve growth and disease resistance of cultured fish. Our study aimed to investigate the effect of dietary stabilized culture of Lactobacillus rhamnosus GCC-3 on growth performance, gut and liver health and anti-viral ability of zebrafish (Danio rerio). Zebrafish (0.161 ± 0.001 g) were fed control and the experimental diet containing 1% GCC-3 culture (1 × 107 CFU/g diet) for four weeks. Growth performance and gut and liver health parameters were monitored after four weeks feeding. The gut microbiota was analyzed by 16S rRNA gene sequencing. In another experiment, zebrafish (0.212 ± 0.001 g) were fed with basal or GCC-3 diets and challenged by spring viremia of carp virus (SVCV) at the end of feeding. The antiviral immune response was evaluated at 2nd and 4th days post SVCV infection and survival rate was calculated 14 days after challenge. The results showed that adding 1% GCC-3 significantly improved growth performance of zebrafish (P < 0.05). The intestinal expression of hypoxia-inducible factor Hif-1α, tight junction protein ZO-1α and ZO-1β was significantly up-regulated in 1% GCC-3 group compared with control (P < 0.05). Besides, 1% GCC-3 decreased the content of MDA and increased total antioxidant capacity in the intestine, and the relative expression of SOD, GST and Gpxa was improved. The abundance of Proteobacteria was reduced while Firmicutes was enriched in the intestinal microbiota of 1% GCC-3 group compared with control (P < 0.05). Zebrafish fed 1% GCC-3 showed higher survival rate after SVCV challenge. Accordingly, the expression of antiviral genes in the spleen was increased at 2nd and 4th days post infection. In conclusion, our results indicate that dietary 1% GCC-3 supplementation can improve gut and liver health as well as antiviral immunity of zebrafish.
Collapse
Affiliation(s)
- Hui Liang
- Sino-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Yadong Xie
- Sino-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Ming Li
- Sino-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Jie Chen
- Sino-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Wenhao Zhou
- Sino-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Rui Xia
- Sino-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Qianwen Ding
- Norway-China Joint Lab on Fish Gastrointestinal Microbiota, Institute of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Yuanyuan Yao
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Zhen Zhang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Yalin Yang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Chao Ran
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| | - Zhigang Zhou
- Sino-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| |
Collapse
|
10
|
Hou Y, Michiels J, Kerschaver CV, Vandaele M, Majdeddin M, Vossen E, Degroote J. The kinetics of glutathione in the gastrointestinal tract of weaned piglets supplemented with different doses of dietary reduced glutathione. Front Vet Sci 2023; 10:1220213. [PMID: 37635757 PMCID: PMC10448897 DOI: 10.3389/fvets.2023.1220213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/15/2023] [Accepted: 07/17/2023] [Indexed: 08/29/2023] Open
Abstract
This study aimed to investigate the kinetics of dietary GSH in the gastrointestinal tract and the effect of GSH on the intestinal redox status of weaned piglets. Forty-eight piglets with an average age of 26 days and an average body weight of 7.7 kg were used in this study. The piglets were divided into three treatment groups including the control group with a basal diet (CON) and two GSH groups with a basal diet supplemented with 0.1% GSH (LGSH) and 1.0% GSH (HGSH), respectively. The basal diet did not contain any GSH. The experiment lasted for 14 days, with eight animals sampled from each group on d5 and 14. The parts of 0-5%, 5-75%, and 75-100% of the length of the small intestine were assigned to SI1, SI2, and SI3. The results showed that GSH almost completely disappeared from the digesta at SI2. However, no difference in the GSH level in mucosa, liver, and blood erythrocytes was found. The level of cysteine (CYS) in SI1 digesta was significantly higher in HGSH than CON and LGSH on d14, and similar findings were observed for cystine (CYSS) in SI3 digesta on d5. The CYSS level in HGSH was also significantly higher than LGSH in the stomach on d14, while no CYS or CYSS was detected in the stomach for control animals, indicating the breakdown of GSH to CYS already occurred in the stomach. Irrespective of the dietary treatment, the CYS level on d14 and the CYSS level on d5 and 14 were increased when moving more distally into the gastrointestinal tract. Furthermore, the mucosal CYS level was significantly increased at SI1 in the LGSH and HGSH group compared with CON on d5. Glutathione disulfide (GSSG) was recovered in the diets and digesta from the LGSH and HGSH group, which could demonstrate the auto-oxidation of GSH. It is, therefore, concluded that GSH supplementation could not increase the small intestinal mucosal GSH level of weaned piglets, and this could potentially relate to the kinetics of GSH in the digestive tract, where GSH seemed to be prone to the breakdown to CYS and CYSS and the auto-oxidation to GSSG.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jeroen Degroote
- Laboratory of Animal Nutrition and Animal Product Quality (LANUPRO), Department of Animal Sciences and Aquatic Ecology, Ghent University, Ghent, Belgium
| |
Collapse
|
11
|
Wei JD, Xu X. Oxidative stress in Wernicke's encephalopathy. Front Aging Neurosci 2023; 15:1150878. [PMID: 37261263 PMCID: PMC10229051 DOI: 10.3389/fnagi.2023.1150878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/25/2023] [Accepted: 05/02/2023] [Indexed: 06/02/2023] Open
Abstract
Wernicke's encephalopathy (WE) is a severe life-threatening disease that occurs due to vitamin B1 (thiamine) deficiency (TD). It is characterized by acute mental disorder, ataxia, and ophthalmoplegia. TD occurs because of the following reasons: insufficient intake, increased demand, and long-term drinking due to corresponding organ damage or failure. Recent studies showed that oxidative stress (OS) can damage organs and cause TD in the brain, which further leads to neurodegenerative diseases, such as WE. In this review, we discuss the effects of TD caused by OS on multiple organ systems, including the liver, intestines, and brain in WE. We believe that strengthening the human antioxidant system and reducing TD can effectively treat WE.
Collapse
Affiliation(s)
- Jun-Dong Wei
- Department of Basic Medical Science, Medical College, Taizhou University, Taizhou, China
| | - Xueming Xu
- Department of Psychiatry, Taizhou Second People's Hospital, Taizhou, China
| |
Collapse
|
12
|
Chen L, Zhu M, Liu Y, Yang Z, Li H, Mu H, Liu S, Wu B. Perfluorobutanesulfonate exposure induces metabolic disturbances in different regions of mouse gut. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 866:161261. [PMID: 36587682 DOI: 10.1016/j.scitotenv.2022.161261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 10/21/2022] [Revised: 12/24/2022] [Accepted: 12/24/2022] [Indexed: 06/17/2023]
Abstract
Perfluorobutanesulfonate (PFBS), an alternative to perfluorooctanesulfonate (PFOS), has raised many health concerns. However, PFBS toxicity in the mammalian gut remains unclear. C57BL/6 mice were exposed to 10 μg/L and 500 μg/L PFBS or 500 μg/L PFOS in their water supply for 28 days. PFBS toxicity in the ileum and colon was explored and compared to that of PFOS. Biochemical analysis showed that tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) levels increased in the ileum exposed to 10 μg/L PFBS, whereas no significant changes were observed in those levels in the colon. Catalase (CAT) activity, malondialdehyde (MDA), TNF-α, and IL-1β levels increased and glutathione (GSH) levels decreased in the ileum of the 500 μg/L-PFBS group, whereas only MDA levels increased in the colon of the 500 μg/L-PFBS group. The results showed that more severe damage occurred in the ileum than in the colon after PFBS exposure, and these align with the 500 μg/L-PFOS group exposure as well. Furthermore, metabolomic analysis revealed glutathione metabolism as a vital factor in inducing PFBS and PFOS toxicities in the ileum. Steroid hormone and amino acid metabolisms were other important factors involved in PFBS and PFOS toxicities, respectively. In the colon, GSH, pyrimidine, and glucose (especially galactose) metabolism was the main contributor to PFBS toxicity, and sulfur amino acid metabolism was the main pathway for PFOS toxicity. This study provides more evidence of the health hazards due to low-dose PFBS exposure in the mammalian gut.
Collapse
Affiliation(s)
- Ling Chen
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, PR China
| | - Mengyuan Zhu
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, PR China
| | - Yafeng Liu
- Department of Environmental Science, School of Engineering, China Pharmaceutical University, Nanjing 211198, PR China
| | - Zhongchao Yang
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, PR China
| | - Huan Li
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, PR China
| | - Hongxin Mu
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, PR China
| | - Su Liu
- Department of Environmental Science, School of Engineering, China Pharmaceutical University, Nanjing 211198, PR China; School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, PR China
| | - Bing Wu
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, PR China.
| |
Collapse
|
13
|
Wang H, Ainiwaer A, Song Y, Qin L, Peng A, Bao H, Qin H. Perturbed gut microbiome and fecal and serum metabolomes are associated with chronic kidney disease severity. MICROBIOME 2023; 11:3. [PMID: 36624472 PMCID: PMC9827681 DOI: 10.1186/s40168-022-01443-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Academic Contribution Register] [Received: 10/14/2022] [Accepted: 12/02/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Chronic kidney disease (CKD) is a severe public health problem associated with a disordered gut microbiome. However, the functional alterations of microbiota and their cross talk with metabolism pathways based on disease severity remain unclear. RESULTS We performed metagenomics and untargeted metabolomics in a cohort of 68 patients with CKD of differing severities and 20 healthy controls to characterize the complex interplay between the gut microbiome and fecal and serum metabolites during CKD progression. We identified 26 microbial species that significantly changed in patients with CKD; 18 species changed as the disease progressed, and eight species changed only in a specific CKD group. These distinct changes in gut microbiota were accompanied by functional alterations in arginine and proline, arachidonic acid, and glutathione metabolism and ubiquinone and other terpenoid-quinone biosynthesis pathways during CKD progression. Further metabolomic analyses revealed that the distributions of toxic and pro-oxidant metabolites from these four essential metabolic pathways varied in the feces and serum as CKD progressed. Furthermore, we observed a complex co-occurrence between CKD severity-related bacteria and the characterized metabolites from the four essential metabolic pathways. Notably, Ruminococcus bromii, fecal hydroquinone, and serum creatinine were identified as the main contributors to the integrated network, indicating their key roles in CKD progression. Moreover, a noninvasive model including R. bromii and fecal hydroquinone, L-cystine, and 12-keto-tetrahydro-LTB4 levels classified the CKD severity (area under the curve [AUC]: > 0.9) and had better performance than the serum creatinine level for mild CKD (AUC: 0.972 vs. 0.896). CONCLUSIONS Perturbed CKD severity-related gut microbiota may contribute to unbalanced toxic and pro-oxidant metabolism in the gut and host, accelerating CKD progression, which may be an early diagnostic and therapeutic target for CKD. Video Abstract.
Collapse
Affiliation(s)
- Haichao Wang
- Department of Nephrology and Rheumatology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Aisima Ainiwaer
- Department of Nephrology and Rheumatology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Yaxiang Song
- Department of Nephrology and Rheumatology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Ling Qin
- Department of Nephrology and Rheumatology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Ai Peng
- Department of Nephrology and Rheumatology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Hui Bao
- Department of Nephrology and Rheumatology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Huanlong Qin
- Department of Gastrointestinal Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| |
Collapse
|
14
|
Li W, He E, Zhang P, Li Y, Qiu H. Multiomics analyses uncover nanoceria triggered oxidative injury and nutrient imbalance in earthworm Eisenia fetida. JOURNAL OF HAZARDOUS MATERIALS 2022; 437:129354. [PMID: 35717815 DOI: 10.1016/j.jhazmat.2022.129354] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 02/15/2022] [Revised: 05/24/2022] [Accepted: 06/08/2022] [Indexed: 06/15/2023]
Abstract
The toxic stress caused by nanoceria remains vague owing to the limited efforts scrutinizing its molecular mechanisms. Herein, we investigated the impacts of nanoceria on earthworm Eisenia fetida, at the molecular level using the multiomics-based profiling approaches (transcriptomics, metabolomics, and 16 S rRNA sequencing). Nanoceria (50 and 500 mg/kg) significantly increased the contents of malondialdehyde (MDA), Fe, and K in worms, suggesting oxidative injury and nutrient imbalance. This was corroborated by the transcriptomic and metabolomic analyses. Nanoceria decreased the levels of certain genes and metabolites associated with glycerolipid and glycerophospholipid metabolisms, suggesting the production of reactive oxygen species and subsequent oxidative stress. Additionally, the ABCD3 gene belonging to ABC transporter family was upregulated, facilitating Fe uptake by worms. Moreover, the higher contents of MDA, Fe, and K after exposure were tightly associated with the imbalanced intestinal flora. Specifically, a higher relative abundance of Actinobacteriota and a lower relative abundance of Proteobacteria and Patescibacteria were induced. This study, for the first time, revealed that nanoceria at nonlethal levels caused oxidative stress and nutrient imbalance of earthworms from the perspective of genes, metabolites, and gut microbiome perturbations, and also established links between the gut microbiome and the overall physiological responses of the host.
Collapse
Affiliation(s)
- Wenxing Li
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Erkai He
- School of Geographic Sciences, East China Normal University, Shanghai 200241, China
| | - Peihua Zhang
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yinsheng Li
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hao Qiu
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
15
|
Tian S, Wang J, Gao R, Wang J, Zhu W. Early-life galacto-oligosaccharides supplementation alleviates the small intestinal oxidative stress and dysfunction of lipopolysaccharide-challenged suckling piglets. J Anim Sci Biotechnol 2022; 13:70. [PMID: 35655292 PMCID: PMC9164537 DOI: 10.1186/s40104-022-00711-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/04/2021] [Accepted: 04/01/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Galacto-oligosaccharides (GOS) are non-digestible food ingredients that promote the growth of beneficial bacteria in the gut. This study investigated the protective effect of the early-life GOS supplement on the piglets' gut function against the oxidative stress induced by lipopolysaccharide (LPS)-challenge. METHODS Eighteen neonatal piglets were assigned to three groups including CON, LPS and LPS + GOS groups. The piglets in CON group and LPS group received physiological saline, while those in LPS + GOS group received GOS solution for 13 d after birth. On d 14, the piglets in LPS group and LPS + GOS group were injected with LPS solutions, while the piglets in CON group were injected with the same volume of physiological saline. RESULTS The results showed that the early-life GOS supplement blocked the LPS-induced reactive oxygen species (ROS) secretion, malondialdehyde (MDA) production and the increase of pro-apoptotic factor expression. Meanwhile, the early-life GOS supplement improved the activities of antioxidant enzymes, disaccharidase enzymes activities, and digestive enzymes activities, and increased the mRNA abundance of the gene related to nutrient digestion and absorption and the relative protein expression of tight junction. The study also showed that the early-life GOS supplement improved the expression of Hemeoxygenase-1 (HO-1) and NAD(P)H/quinone acceptor oxidoreductase-1 (NQO-1), and activated the AMP-activated protein kinase (AMPK). CONCLUSIONS These results suggested that GOS enhanced the gut function, reduced the ROS production and pro-apoptotic factors gene expression, and activated the AMPK signaling pathway in LPS-challenged piglets.
Collapse
Affiliation(s)
- Shiyi Tian
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jue Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ren Gao
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jing Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
16
|
The Intestinal Redox System and Its Significance in Chemotherapy-Induced Intestinal Mucositis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7255497. [PMID: 35585883 PMCID: PMC9110227 DOI: 10.1155/2022/7255497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 03/02/2022] [Revised: 04/04/2022] [Accepted: 04/09/2022] [Indexed: 12/12/2022]
Abstract
Chemotherapy-induced intestinal mucositis (CIM) is a significant dose-limiting adverse reaction brought on by the cancer treatment. Multiple studies reported that reactive oxygen species (ROS) is rapidly produced during the initial stages of chemotherapy, when the drugs elicit direct damage to intestinal mucosal cells, which, in turn, results in necrosis, mitochondrial dysfunction, and ROS production. However, the mechanism behind the intestinal redox system-based induction of intestinal mucosal injury and necrosis of CIM is still undetermined. In this article, we summarized relevant information regarding the intestinal redox system, including the composition and regulation of redox enzymes, ROS generation, and its regulation in the intestine. We innovatively proposed the intestinal redox “Tai Chi” theory and revealed its significance in the pathogenesis of CIM. We also conducted an extensive review of the English language-based literatures involving oxidative stress (OS) and its involvement in the pathological mechanisms of CIM. From the date of inception till July 31, 2021, 51 related articles were selected. Based on our analysis of these articles, only five chemotherapeutic drugs, namely, MTX, 5-FU, cisplatin, CPT-11, and oxaliplatin were shown to trigger the ROS-based pathological mechanisms of CIM. We also discussed the redox system-mediated modulation of CIM pathogenesis via elaboration of the relationship between chemotherapeutic drugs and the redox system. It is our belief that this overview of the intestinal redox system and its role in CIM pathogenesis will greatly enhance research direction and improve CIM management in the future.
Collapse
|
17
|
Dore MP, Errigo A, Bibbò S, Manca A, Pes GM. High Frequency of Glucose-6-Phosphate Dehydrogenase Deficiency in Patients Diagnosed with Celiac Disease. Nutrients 2022; 14:1815. [PMID: 35565779 PMCID: PMC9099929 DOI: 10.3390/nu14091815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/19/2022] [Revised: 04/14/2022] [Accepted: 04/25/2022] [Indexed: 01/13/2023] Open
Abstract
Celiac disease (CD) is characterized by a proinflammatory state associated with the production of reactive oxygen species, i.e., a condition of oxidative stress. In this study, we tested the hypothesis that the inherited deficiency of glucose-6-phosphate dehydrogenase (G6PD), by causing impaired antioxidant defense, may increase the risk of CD. METHODS A retrospective monocentric case-control study was performed using the clinical records of 8338 outpatients (64.6% women) scheduled for upper endoscopy between 2002 and 2021 in Northern Sardinia. Overall, 627 were found to have CD (7.5%), and 1027 resulted to be G6PD-deficiency carriers (12.3%). Since randomization was impractical, the potential covariates imbalance between cases and controls was minimized using a 1:2 propensity-score-matched (PSM) analysis. RESULTS Overall, G6PD deficiency was associated with increased risk of CD (odds ratio (OR) 1.50; 95% confidence interval (CI) 1.19-1.90). The PSM procedure identified 1027 G6PD-deficient and 2054 normal patients. Logistic regression including the propensity score detected for G6PD deficiency an OR of 1.48 (95%CI 1.13-1.95; p = 0.004). CONCLUSIONS Our findings show that the enzyme defect was significantly and positively associated with CD, in line with the pro-oxidant impact of the enzyme defect observed in animal models and humans.
Collapse
Affiliation(s)
- Maria Pina Dore
- Dipartimento di Medicina, Chirurgia e Farmacia, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy; (S.B.); (A.M.); (G.M.P.)
- Baylor College of Medicine, One Baylor Plaza Blvd, Houston, TX 77030, USA
| | - Alessandra Errigo
- Dipartimento di Scienze Biomediche, University of Sassari, Viale San Pietro 43B, 07100 Sassari, Italy;
| | - Stefano Bibbò
- Dipartimento di Medicina, Chirurgia e Farmacia, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy; (S.B.); (A.M.); (G.M.P.)
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Roma, Italy
| | - Alessandra Manca
- Dipartimento di Medicina, Chirurgia e Farmacia, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy; (S.B.); (A.M.); (G.M.P.)
| | - Giovanni Mario Pes
- Dipartimento di Medicina, Chirurgia e Farmacia, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy; (S.B.); (A.M.); (G.M.P.)
- Sardinia Longevity Blue Zone Observatory, 08040 Nuoro, Italy
| |
Collapse
|
18
|
Cui Y, Zhou X, Chen L, Tang Z, Mo F, Li XC, Mao H, Wei X, Wang C, Wang H. Crosstalk between Endoplasmic Reticulum Stress and Oxidative Stress in Heat Exposure-Induced Apoptosis Is Dependent on the ATF4-CHOP-CHAC1 Signal Pathway in IPEC-J2 Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:15495-15511. [PMID: 34919378 DOI: 10.1021/acs.jafc.1c03361] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 06/14/2023]
Abstract
The intestinal epithelium is susceptible to heat stress (HS), which leads to gut leakage and inflammation. However, the mechanisms underlying HS-induced intestine dysfunction have yet to be elucidated. We established an in vitro chronic heat exposure-induced intestinal injury of intestinal porcine epithelial cells (IPEC-J2) exposed to high temperatures (43 °C) for 12 h. The results revealed that HS increased reactive oxygen species (ROS) generation and decreased superoxide dismutase 2 (SOD2) expression, leading to oxidative stress. Western blotting analysis demonstrated that HS induced apoptosis as evidenced by increased cytochrome c (Cyt c) release in the cytoplasm and caspase 3 activation. Transcriptome sequencing analysis revealed that HS activated the endoplasmic reticulum stress (ERS) response/unfolded protein response (UPR) but inhibited glutathione metabolism. Specifically, HS triggered the pro-apoptotic activating transcription factor 4 (ATF4)/CEBP-homologous protein (CHOP) branch of the UPR. Interestingly, glutathione-specific gamma-glutamylcyclotransferase1 (CHAC1) involved in glutathione degradation was upregulated due to heat exposure and was proved to be downstream of the ATF4-CHOP signal pathway. Knockdown of CHAC1 attenuated the HS-induced decrease in glutathione level and cell apoptosis. These studies suggest that crosstalk between ERS and oxidative stress in HS-induced apoptosis might be dependent on the ATF4-CHOP-CHAC1 signal pathway in IPEC-J2 cells.
Collapse
Affiliation(s)
- Yanjun Cui
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Lin'an 311300, P. R. China
| | - Xu Zhou
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Lin'an 311300, P. R. China
| | - Leyi Chen
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Lin'an 311300, P. R. China
| | - Zhining Tang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Lin'an 311300, P. R. China
| | - Fan Mo
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Lin'an 311300, P. R. China
| | - Xiang Chen Li
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Lin'an 311300, P. R. China
| | - Huiling Mao
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Lin'an 311300, P. R. China
| | - Xiaoshi Wei
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Lin'an 311300, P. R. China
| | - Chong Wang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Lin'an 311300, P. R. China
| | - Haifeng Wang
- College of Animal Science, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310058, P. R. China
| |
Collapse
|
19
|
Al-Gubory KH, Laher I, Garrel C. Pomegranate peel attenuates dextran sulfate sodium-induced lipid peroxidation in rat small intestine by enhancing the glutathione/glutathione disulfide redox potential. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:4278-4287. [PMID: 33417238 DOI: 10.1002/jsfa.11067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 10/14/2020] [Revised: 01/03/2021] [Accepted: 01/08/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND The peel of the pomegranate fruit is rich in polyphenols with antioxidant properties. We investigated the preventive effect of pomegranate peel (PP) powder against dextran sulfate sodium (DSS)-induced lipid peroxidation in the small intestine of rats. Rats were allocated to four groups: CONT group, fed a standard rodent diet; DSS group, fed a standard rodent diet and treated with DSS; as well as PP1%+DSS and PP5%+DSS groups, fed a standard rodent diet supplemented with either 1% or 5% of PP powder and treated with DSS. Rats of the four groups consumed their diets for 25 days. Lipid peroxidation was determined by measuring malondialdehyde (MDA) concentrations in plasma and MDA contents in the small intestine and liver. Glutathione/glutathione disulfide (GSH/GSSG) redox status and antioxidant enzyme activities were determined in the small intestine and liver. RESULTS MDA content was higher (P < 0.001) in the small intestines of the DSS group compared to the CONT group. MDA content was reduced (P < 0.001) in the small intestines of the PP1%+DSS and PP5%+DSS groups compared to the DSS group. GSH contents and GSH/GSSG ratios were higher (P < 0.001) in the small intestines of the PP5%+DSS group compared to the CONT, DSS and PP1%+DSS groups. CONCLUSION The present study demonstrates that PP powder protects the small intestine against DSS-induced lipid peroxidation by enhancing the GSH/GSSG redox potential. Powder of PP is a promising agricultural by-product containing a mixture of bioactive polyphenols that can be used for the production of functional foods aimed at the prevention of oxidative stress-induced small intestine pathogenesis. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Kaïs H Al-Gubory
- Department of Animal Physiology and Livestock Systems, National Institute for Agriculture, Food and Environment, 78352 Jouy-en-Josas Cedex, France
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Catherine Garrel
- Department of Biology, Toxicology and Pharmacology, Nutritional and Hormonal Biochemistry Unit, Institute of Biology and Pathology, University Hospital Center of Grenoble, 38043 Grenoble Cedex 9, France
| |
Collapse
|
20
|
Zhang S, Wang C, Zhong W, Kemp AH, Guo M, Killpartrick A. Polymerized Whey Protein Concentrate-Based Glutathione Delivery System: Physicochemical Characterization, Bioavailability and Sub-Chronic Toxicity Evaluation. Molecules 2021; 26:1824. [PMID: 33805036 PMCID: PMC8037743 DOI: 10.3390/molecules26071824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/21/2021] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 11/20/2022] Open
Abstract
Glutathione (GSH) is a powerful antioxidant, but its application is limited due to poor storage stability and low bioavailability. A novel nutrient encapsulation and delivery system, consisting of polymerized whey protein concentrate and GSH, was prepared and in vivo bioavailability, antioxidant capacity and toxicity were evaluated. Polymerized whey protein concentrate encapsulated GSH (PWPC-GSH) showed a diameter of roughly 1115 ± 7.07 nm (D50) and zeta potential of 30.37 ± 0.75 mV. Differential scanning calorimetry (DSC) confirmed that GSH was successfully dispersed in PWPC particles. In vivo pharmacokinetics study suggested that PWPC-GSH displayed 2.5-times and 2.6-fold enhancement in maximum concentration (Cmax) and area under the concentration-time curve (AUC) as compared to free GSH. Additionally, compared with plasma of mice gavage with free GSH, significantly increased antioxidant capacity of plasma in mice with PWPC-GSH was observed (p < 0.05). Sub-chronic toxicity evaluation indicated that no adverse toxicological reactions related to oral administration of PWPC-GSH were observed on male and female rats with a diet containing PWPC-GSH up to 4% (w/w). Data indicated that PWPC may be an effective carrier for GSH to improve bioavailability and antioxidant capacity.
Collapse
Affiliation(s)
- Siyu Zhang
- Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (S.Z.); (M.G.)
| | - Cuina Wang
- Department of Food Science, Jilin University, Changchun 130000, China; (C.W.); (W.Z.)
| | - Weigang Zhong
- Department of Food Science, Jilin University, Changchun 130000, China; (C.W.); (W.Z.)
| | - Alyssa H. Kemp
- Department of Nutrition and Food Sciences, University of Vermont, Burlington, VT 05403, USA;
| | - Mingruo Guo
- Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (S.Z.); (M.G.)
- Department of Nutrition and Food Sciences, University of Vermont, Burlington, VT 05403, USA;
| | - Adam Killpartrick
- Department of Nutrition and Food Sciences, University of Vermont, Burlington, VT 05403, USA;
| |
Collapse
|
21
|
Wang Y, Chen Y, Zhang X, Lu Y, Chen H. New insights in intestinal oxidative stress damage and the health intervention effects of nutrients: A review. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104248] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/06/2023] Open
|
22
|
Xiong L, You J, Zhang W, Zhu Q, Blachier F, Yin Y, Kong X. Intrauterine growth restriction alters growth performance, plasma hormones, and small intestinal microbial communities in growing-finishing pigs. J Anim Sci Biotechnol 2020; 11:86. [PMID: 32832077 PMCID: PMC7437023 DOI: 10.1186/s40104-020-00490-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/05/2020] [Accepted: 06/23/2020] [Indexed: 12/16/2022] Open
Abstract
Background The interaction of the gut microbiota with key metabolic and physiological processes may be associated with poor growth outcomes in animals born with intrauterine growth restriction (IUGR). Results Growth performance, plasma hormone concentrations, and intestinal microbiota composition were analyzed in IUGR pigs and in normal birth weight (NBW) pigs when the NBW pigs reached 25, 50, and 100 kg of body weight (BW). Compared to NBW pigs, IUGR pigs had lower initial, weaned, and final BW, and lower average daily gain and average daily feed intake in all the considered time points. In the 25 kg BW group, IUGR pigs had higher concentrations of plasma ghrelin and pancreatic polypeptide (PP), but lower insulin concentration than NBW pigs, while the situation was reversed in the 50 kg BW group. As compared to NBW pigs, IUGR pigs had higher microbial alpha diversity in the jejunum and ileum; in the 50 and 100 kg BW groups, IUGR pigs had higher Firmicutes abundance but lower Proteobacteria abundance in the jejunum, and lower Lactobacillus abundance in the jejunum and ileum; in the 25 kg BW group, IUGR pigs showed higher unclassified Ruminococcaceae abundance in the ileum; and in 25 and 50 kg BW groups, IUGR pigs showed lower Ochrobactrum abundance in the jejunum. Spearman’s correlation revealed that Lactobacillus was negatively correlated with growth performance, while unclassified Ruminococcaceae was positively correlated. Predictive metagenomic analysis detected significantly different expression of genes in the intestinal microbiota between IUGR and NBW pigs, suggesting different metabolic capabilities between the two groups. Conclusions Growing-finishing IUGR pigs showed lower growth performance, higher microbial alpha diversity, and differences in plasma hormone concentrations compared to NBW pigs. Alterations in the abundance of Firmicutes, Proteobacteria, Ruminococcaceae, Lactobacillus, and Ochrobactrum in the small intestine may be associated with IUGR, and may therefore serve as a future target for gut microbiota intervention in growing-finishing IUGR pigs.
Collapse
Affiliation(s)
- Liang Xiong
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Regions, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125 Hunan China.,Key Laboratory of Animal Nutrition in Jiangxi Province, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 440000 Jiangxi China
| | - Jinming You
- Key Laboratory of Animal Nutrition in Jiangxi Province, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 440000 Jiangxi China
| | - Wanghong Zhang
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Regions, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125 Hunan China
| | - Qian Zhu
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Regions, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125 Hunan China
| | - Francois Blachier
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 75005 Paris, France
| | - Yulong Yin
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Regions, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125 Hunan China
| | - Xiangfeng Kong
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Regions, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125 Hunan China
| |
Collapse
|
23
|
Wolfarth AA, Liu X, Darby TM, Boyer DJ, Spizman JB, Owens JA, Chandrasekharan B, Naudin CR, Hanley KZ, Robinson BS, Ortlund EA, Jones RM, Neish AS. Proline-Rich Acidic Protein 1 (PRAP1) Protects the Gastrointestinal Epithelium From Irradiation-Induced Apoptosis. Cell Mol Gastroenterol Hepatol 2020; 10:713-727. [PMID: 32629119 PMCID: PMC7498829 DOI: 10.1016/j.jcmgh.2020.06.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/16/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS The intestinal epithelium must be resilient to physiochemical stress to uphold the physiological barrier separating the systemic compartment from the microbial and antigenic components of the gut lumen. Identifying proteins that mediate protection and enhancing their expression is therefore a clear approach to promote intestinal health. We previously reported that oral ingestion of the probiotic Lactobacillus rhamnosus GG not only induced the expression of several recognized cytoprotective factors in the murine colon, but also many genes with no previously described function, including the gene encoding proline-rich acidic protein 1 (PRAP1). PRAP1 is a highly expressed protein in the epithelium of the gastrointestinal tract and we sought to define its function in this tissue. METHODS Purified preparations of recombinant PRAP1 were analyzed biochemically and PRAP1 antisera were used to visualize localization in tissues. Prap1-/- mice were characterized at baseline and challenged with total body irradiation, then enteroids were generated to recapitulate the irradiation challenge ex vivo. RESULTS PRAP1 is a 17-kilodalton intrinsically disordered protein with no recognizable sequence homology. PRAP1 expression levels were high in the epithelia of the small intestine. Although Prap1-/- mice presented only mild phenotypes at baseline, they were highly susceptible to intestinal injury upon challenge. After irradiation, the Prap1-/- mice showed accelerated death with a significant increase in apoptosis and p21 expression in the small intestinal epithelium. CONCLUSIONS PRAP1 is an intrinsically disordered protein highly expressed by the gastrointestinal epithelium and functions at exposed surfaces to protect the barrier from oxidative insult.
Collapse
Affiliation(s)
- Alexandra A Wolfarth
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Xu Liu
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia
| | - Trevor M Darby
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Darra J Boyer
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Jocelyn B Spizman
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Joshua A Owens
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Bindu Chandrasekharan
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Crystal R Naudin
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Krisztina Z Hanley
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Brian S Robinson
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Eric A Ortlund
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia
| | - Rheinallt M Jones
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia; Emory Microbiome Research Center, Emory University School of Medicine, Atlanta, Georgia
| | - Andrew S Neish
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia; Emory Microbiome Research Center, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
24
|
Dong Y, Hou Q, Lei J, Wolf PG, Ayansola H, Zhang B. Quercetin Alleviates Intestinal Oxidative Damage Induced by H 2O 2 via Modulation of GSH: In Vitro Screening and In Vivo Evaluation in a Colitis Model of Mice. ACS OMEGA 2020; 5:8334-8346. [PMID: 32309744 PMCID: PMC7161027 DOI: 10.1021/acsomega.0c00804] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 02/23/2020] [Accepted: 03/24/2020] [Indexed: 05/13/2023]
Abstract
The gastrointestinal tract is exposed to pro-oxidants from food, host immune factors, and microbial pathogens, which may induce oxidative damage. Oxidative stress has been shown to play an important role in the onset of inflammatory bowel disease. This study aimed to use a novel model to evaluate the effects of a screened natural component and explore its possible mechanism. An in vitro oxidative stress Caco2 cell model induced by H2O2 was established using a real-time cellular analysis system and verified by addition of glutathione (GSH). A variety of plant components were chosen for the screening. Quercetin was the most effective phytochemical to alleviate the decreased cell index caused by H2O2 among the tested plant components. Furthermore, quercetin ameliorated dextran sulfate sodium salt (DSS)-induced colitis and further increased the serum GSH. The mechanism of quercetin protection was explored in Caco2. Reversed H2O2-induced cell damage and decreased reactive oxygen species and apoptosis ratio were observed in quercetin-treated cells. Also, quercetin increased expression of the glutamate-cysteine ligase catalytic subunit (GCLC), the first rate-limiting enzyme of glutathione synthesis, and increased intracellular GSH concentration under H2O2 treatment. This effect was abolished by the GCLC inhibitor buthionine sulfoximine. These results indicated that quercetin can improve cell proliferation and increase intracellular GSH concentrations by upregulating transcription of GCLC to eliminate excessive reactive oxygen species (ROS). Increased extracellular H2O2 concentration induced by quercetin under oxidative stress was related to the inhibition of AQP3 and upregulation of NOX1/2, which may contribute to the observed protective effects of quercetin. Moreover, the novel H2O2-induced oxidative stress cell model based on the real-time cellular analysis system was an effective model to screen natural products to deal with intestinal oxidative damage and help accelerate the discovery of new drugs for inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Yuanyang Dong
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Qihang Hou
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Jiaqi Lei
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Patricia G. Wolf
- Division
of Nutritional Sciences, University of Illinois
at Urbana-Champaign, 1207 W. Gregory Avenue, Urbana, Illinois 61801, United
States
| | - Hammed Ayansola
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Bingkun Zhang
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
25
|
Liang H, Liu N, Wang R, Zhang Y, Chen J, Dai Z, Yang Y, Wu G, Wu Z. N-Acetyl Serotonin Alleviates Oxidative Damage by Activating Nuclear Factor Erythroid 2-Related Factor 2 Signaling in Porcine Enterocytes. Antioxidants (Basel) 2020; 9:antiox9040303. [PMID: 32272634 PMCID: PMC7222184 DOI: 10.3390/antiox9040303] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/20/2020] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/22/2022] Open
Abstract
Apoptosis of intestinal epithelial cells following oxidative stress is a major cause of mucosal barrier dysfunction and is associated with the pathogenesis of various gastrointestinal diseases. Although L-tryptophan (Trp) is known to improve intestinal integrity and function, a beneficial effect of N-acetyl serotonin (NAS), a metabolite of Trp, on the apoptosis of enterocytes and the underlying mechanisms remain largely unknown. In the present study, we showed that porcine enterocytes treated with 4-hydroxy-2-nonenal (4-HNE), a metabolite of lipid peroxidation, led to upregulation of apoptotic proteins, including Bax and cleaved caspase-3, and reduction of tight junction proteins. These effects of 4-HNE were significantly abrogated by NAS. In addition, NAS reduced ROS accumulation while increasing the intracellular concentration of glutathione (GSH), and the abundance of the Nrf2 protein in the nucleus and its downstream target proteins. Importantly, these protective effects of NAS were abrogated by Atra, an inhibitor of Nrf2, indicating a dependence on Nrf2 signaling. Taken together, we demonstrated that NAS attenuated oxidative stress-induced cellular injury in porcine enterocytes by regulating Nrf2 signaling. These findings provide new insights into a functional role of NAS in maintaining intestinal homeostasis.
Collapse
Affiliation(s)
- Haiwei Liang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.L.); (R.W.); (Y.Z.); (J.C.); (Z.D.); (Y.Y.)
| | - Ning Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China;
| | - Renjie Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.L.); (R.W.); (Y.Z.); (J.C.); (Z.D.); (Y.Y.)
| | - Yunchang Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.L.); (R.W.); (Y.Z.); (J.C.); (Z.D.); (Y.Y.)
| | - Jingqing Chen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.L.); (R.W.); (Y.Z.); (J.C.); (Z.D.); (Y.Y.)
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.L.); (R.W.); (Y.Z.); (J.C.); (Z.D.); (Y.Y.)
| | - Ying Yang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.L.); (R.W.); (Y.Z.); (J.C.); (Z.D.); (Y.Y.)
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA;
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.L.); (R.W.); (Y.Z.); (J.C.); (Z.D.); (Y.Y.)
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
- Correspondence: ; Tel.: +86-10-6273-1003
| |
Collapse
|
26
|
Liu XW, Feng L, Jiang WD, Wu P, Yang DM, Tang L, Kuang SY, Shi HQ, Zhou XQ, Liu Y. Novel insights into the intestinal immune regulatory effects of (2-Carboxyethyl) dimethylsulfonium Bromide (Br-DMPT) in on-growing grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2020; 98:534-550. [PMID: 32004614 DOI: 10.1016/j.fsi.2020.01.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 10/13/2019] [Revised: 01/05/2020] [Accepted: 01/27/2020] [Indexed: 06/10/2023]
Abstract
The present study evaluated the effects of (2-Carboxyethyl)dimethylsulfonium Bromide (Br-DMPT) supplementation on the intestinal immune function and potential mechanisms of on-growing grass carp (Ctenopharyngodon idella) by feeding fish (initial weight 216.49 ± 0.29 g) five diets with gradational Br-DMPT (0-520 mg/kg diet) concentrations for 60 days and then infecting them with Aeromonas hydrophila for 14 days. Our results firstly indicated that compared with the control group, appropriate Br-DMPT supplementation increased the number of beneficial bacteria Lactobacillus and Bifidobacterium and enteritis resistance, decreased the number of detrimental bacteria Aeromonas and E. coli, and relieved the intestinal histopathological symptoms of fish. In addition, compared with the control group, appropriate Br-DMPT supplementation (1) increased lysozyme (LZ) and acid phosphatase (ACP) activities, as well as complement 3 (C3), C4 and immunoglobulin M (IgM) content; (2) upregulated the mRNA levels of anti-microbial substance: liver expressed anti-microbial peptide (LEAP) -2A, LEAP-2B, hepcidin, β-defensin-1 and Mucin2; (3) partially downregulated the mRNA levels of pro-inflammatory cytokines [interleukin 1β (IL-1β), IL-6, IL-8, IL-12p40, IL-15, IL-17D, tumour necrosis factor α (TNF-α) and interferon γ2 (IFN-γ2)] by inhibiting [IKKβ/IκBα/(NF-κBp65 and c-Rel)] signalling; and (4) partially upregulated the mRNA levels of anti-inflammatory cytokines [IL-4/13A, IL-10, IL-11, transforming growth factor (TGF)-β1] by activating [TOR/(S6K1 and 4E-BP)] signalling. The aforementioned results indicated that appropriate amount of Br-DMPT exerted a positive effect on the regulation of intestinal immune function in fish. Finally, based on enteritis morbidity, the IgM content and the lysozyme activity in the PI, the appropriate levels of Br-DMPT supplementation for on-growing grass carp were established as 295.43, 301.73 and 320.36 mg/kg diet, respectively.
Collapse
Affiliation(s)
- Xing-Wei Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Fish Nutrition and Safety in Production Sichuan University Key Laboratory, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Province, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Fish Nutrition and Safety in Production Sichuan University Key Laboratory, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Fish Nutrition and Safety in Production Sichuan University Key Laboratory, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, China
| | - De-Min Yang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu, 610066, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu, 610066, China
| | - He-Qun Shi
- Guangzhou Cohoo Biotech Co., Ltd., Guangzhou, 510663, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Fish Nutrition and Safety in Production Sichuan University Key Laboratory, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Province, China.
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Fish Nutrition and Safety in Production Sichuan University Key Laboratory, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease-resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, China.
| |
Collapse
|
27
|
Okkelman IA, Neto N, Papkovsky DB, Monaghan MG, Dmitriev RI. A deeper understanding of intestinal organoid metabolism revealed by combining fluorescence lifetime imaging microscopy (FLIM) and extracellular flux analyses. Redox Biol 2020; 30:101420. [PMID: 31935648 PMCID: PMC6957829 DOI: 10.1016/j.redox.2019.101420] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/25/2019] [Revised: 12/13/2019] [Accepted: 12/29/2019] [Indexed: 12/21/2022] Open
Abstract
Stem cells and the niche in which they reside feature a complex microenvironment with tightly regulated homeostasis, cell-cell interactions and dynamic regulation of metabolism. A significant number of organoid models has been described over the last decade, yet few methodologies can enable single cell level resolution analysis of the stem cell niche metabolic demands, in real-time and without perturbing integrity. Here, we studied the redox metabolism of Lgr5-GFP intestinal organoids by two emerging microscopy approaches based on luminescence lifetime measurement - fluorescence-based FLIM for NAD(P)H, and phosphorescence-based PLIM for real-time oxygenation. We found that exposure of stem (Lgr5-GFP) and differentiated (no GFP) cells to high and low glucose concentrations resulted in measurable shifts in oxygenation and redox status. NAD(P)H-FLIM and O2-PLIM both indicated that at high 'basal' glucose conditions, Lgr5-GFP cells had lower activity of oxidative phosphorylation when compared with cells lacking Lgr5. However, when exposed to low (0.5 mM) glucose, stem cells utilized oxidative metabolism more dynamically than non-stem cells. The high heterogeneity of complex 3D architecture and energy production pathways of Lgr5-GFP organoids were also confirmed by the extracellular flux (XF) analysis. Our data reveals that combined analysis of NAD(P)H-FLIM and organoid oxygenation by PLIM represents promising approach for studying stem cell niche metabolism in a live readout.
Collapse
Affiliation(s)
- Irina A Okkelman
- Laboratory of Biophysics and Bioanalysis, ABCRF, University College Cork, Cavanagh Pharmacy Building, College Road, Cork, T12 K8AF, Ireland
| | - Nuno Neto
- Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, Ireland
| | - Dmitri B Papkovsky
- Laboratory of Biophysics and Bioanalysis, ABCRF, University College Cork, Cavanagh Pharmacy Building, College Road, Cork, T12 K8AF, Ireland
| | - Michael G Monaghan
- Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, Ireland; Advanced Materials and BioEngineering Research (AMBER) Centre at Trinity College Dublin and Royal College of Surgeons in Ireland, Dublin, Ireland.
| | - Ruslan I Dmitriev
- Laboratory of Biophysics and Bioanalysis, ABCRF, University College Cork, Cavanagh Pharmacy Building, College Road, Cork, T12 K8AF, Ireland; Institute for Regenerative Medicine, I.M. Sechenov First Moscow State University, Moscow, Russian Federation.
| |
Collapse
|
28
|
Tao S, Zhou T, Saelao P, Wang Y, Zhu Y, Li T, Zhou H, Wang J. Intrauterine Growth Restriction Alters the Genome-Wide DNA Methylation Profiles in Small Intestine, Liver and Longissimus Dorsi Muscle of Newborn Piglets. Curr Protein Pept Sci 2019; 20:713-726. [PMID: 30678618 DOI: 10.2174/1389203720666190124165243] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/12/2018] [Revised: 12/30/2018] [Accepted: 01/01/2019] [Indexed: 01/20/2023]
Abstract
Intrauterine growth restriction (IUGR) remains a major problem in swine production since the associated low birth weight leads to high rates of pre-weaning morbidity and mortality, and permanent retardation of growth and development. The underlying regulatory mechanisms from the aspects of epigenetic modification has received widespread attention. Studies explore the changes in genome wide methylation in small intestine (SI), liver and longissimus dorsi muscle (LDM) between IUGR and normal birth weight (NBW) newborn piglets using a methylated DNA immunoprecipitation-sequencing (MeDIP-Seq) approach. The data demonstrated that methylated peaks were prominently distributed in distal intergenic regions and the quantities of peaks in IUGR piglets were more than that of NBW piglets. IUGR piglets had relatively high methylated level in promoters, introns and coding exons in all the three tissues. Through KEGG pathway analysis of differentially methylated genes found that 33, 54 and 5 differentially methylated genes in small intestine, liver and longissimus dorsi muscle between NBW and IUGR piglets, respectively, which are related to development and differentiation, carbohydrate and energy metabolism, lipid metabolism, protein turnover, immune response, detoxification, oxidative stress and apoptosis pathway. The objective of this review is to assess the impact of differentially methylation status on developmental delay, metabolic disorders and immune deficiency of IUGR piglets.
Collapse
Affiliation(s)
- Shiyu Tao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Tianjiao Zhou
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Perot Saelao
- Department of Animal Science, University of California, Davis, CA 95616, United States
| | - Ying Wang
- Department of Animal Science, University of California, Davis, CA 95616, United States
| | - Yuhua Zhu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Tiantian Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Huaijun Zhou
- Department of Animal Science, University of California, Davis, CA 95616, United States
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
29
|
Fellenberg M, Speisky H. Antioxidants: their effects on broiler oxidative stress and its meat oxidative stability. WORLD POULTRY SCI J 2019. [DOI: 10.1079/wps200584] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/03/2023]
Affiliation(s)
- M.A. Fellenberg
- Departamento de Ciencias Animales, Facultad de Agronomía e Ingeniería Forestal, Pontificia Universidad Católica de Chile. Av. Vicuña Mackenna 4860, Macul, Santiago, Chile
| | - H. Speisky
- Laboratorio de Micronutrientes, Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile
| |
Collapse
|
30
|
Sironi C, Bodega F, Zocchi L, Porta C. Effects of Creatine Treatment on Jejunal Phenotypes in a Rat Model of Acidosis. Antioxidants (Basel) 2019; 8:antiox8070225. [PMID: 31319541 PMCID: PMC6680959 DOI: 10.3390/antiox8070225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/20/2019] [Revised: 07/05/2019] [Accepted: 07/12/2019] [Indexed: 01/03/2023] Open
Abstract
We investigated the effects of creatine treatment on jejunal phenotypes in a rat model of oxidative stress induced by acidosis. In particular, the activities of some antioxidant enzymes (superoxide dismutase, glutathione peroxidase, catalase, and glutathione reductase), the level of lipid peroxidation, the expression of heat shock proteins (HSP70), and the expression of the major carriers of the cells (Na+/K+-ATPase, sodium-glucose Transporter 1—SGLT1, and glucose transporter 2—GLUT2) were measured under control and chronic acidosis conditions. Creatine did not affect the activity of antioxidant enzymes in either the control or acidosis groups, except for catalase, for which the activity was reduced in both conditions. Creatine did not change the lipid peroxidation level or HSP70 expression. Finally, creatine stimulated (Na+/K+)-ATPase expression under both control and chronic acidosis conditions. Chronic acidosis caused reductions in the expression levels of GLUT2 and SGLT1. GLUT2 reduction was abolished by creatine, while the presence of creatine did not induce any strengthening effect on the expression of SGLT1 in either the control or chronic acidosis groups. These results indicate that creatine has antioxidant properties that are realized through direct interaction of the molecule with reactive oxygen species. Moreover, the administration of creatine seems to determine a functional strengthening of the tissue, making it more resistant to acidosis.
Collapse
Affiliation(s)
- Chiara Sironi
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Facoltà di Medicina e Chirurgia, Università degli Studi di Milano, Via Mangiagalli 32, 20133 Milano, Italy
| | - Francesca Bodega
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Facoltà di Medicina e Chirurgia, Università degli Studi di Milano, Via Mangiagalli 32, 20133 Milano, Italy.
| | - Luciano Zocchi
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Facoltà di Medicina e Chirurgia, Università degli Studi di Milano, Via Mangiagalli 32, 20133 Milano, Italy
| | - Cristina Porta
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Facoltà di Medicina e Chirurgia, Università degli Studi di Milano, Via Mangiagalli 32, 20133 Milano, Italy
| |
Collapse
|
31
|
Vissenaekens H, Grootaert C, Rajkovic A, Van De Wiele T, Calatayud M. The response of five intestinal cell lines to anoxic conditionsin vitro. Biol Cell 2019; 111:232-244. [DOI: 10.1111/boc.201800076] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/06/2018] [Revised: 03/22/2019] [Accepted: 05/19/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Hanne Vissenaekens
- Department of Food technologySafety and HealthFaculty of Bioscience EngineeringGhent University Ghent 9000 Belgium
| | - Charlotte Grootaert
- Department of Food technologySafety and HealthFaculty of Bioscience EngineeringGhent University Ghent 9000 Belgium
| | - Andreja Rajkovic
- Department of Food technologySafety and HealthFaculty of Bioscience EngineeringGhent University Ghent 9000 Belgium
| | - Tom Van De Wiele
- Center for Microbial Ecology and Technology (CMET)Faculty of Bioscience EngineeringGhent University Ghent 9000 Belgium
| | - Marta Calatayud
- Center for Microbial Ecology and Technology (CMET)Faculty of Bioscience EngineeringGhent University Ghent 9000 Belgium
| |
Collapse
|
32
|
Huang S, Li N, Liu C, Li T, Wang W, Jiang L, Li Z, Han D, Tao S, Wang J. Characteristics of the gut microbiota colonization, inflammatory profile, and plasma metabolome in intrauterine growth restricted piglets during the first 12 hours after birth. J Microbiol 2019; 57:748-758. [PMID: 31187413 DOI: 10.1007/s12275-019-8690-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/14/2018] [Revised: 02/26/2019] [Accepted: 04/08/2019] [Indexed: 12/13/2022]
Abstract
Intrauterine growth restriction (IUGR) predisposes newborns to inflammatory and metabolic disturbance. Disequilibrium of gut microbiota in early life has been implicated in the incidence of inflammation and metabolic diseases in adulthood. This study aimed to investigate the difference in gut microbiota colonization, cytokines and plasma metabolome between IUGR and normal birth weight (NBW) piglets in early life. At birth, reduced (P < 0.05) body, jejunum, and ileum weights, as well as decreased (P < 0.05) small intestinal villi and increased (P < 0.05) ileal crypt depth were observed in IUGR piglets compared with their NBW counterparts. Imbalanced inflammatory and plasma metabolome profile was observed in IUGR piglets. Furthermore, altered metabolites were mainly involved in fatty acid metabolism and inflammatory response. At 12 h after birth and after suckling colostrum, reduced (P < 0.05) postnatal growth and the small intestinal maturation retardation (P < 0.05) continued in IUGR piglets in comparison with those in NBW littermates. Besides, the gut microbiota structure was significantly altered by IUGR. Importantly, the disruption of the inflammatory profile and metabolic status mainly involved the pro-inflammatory cytokines (IL-1β and IFN-γ) and amino acid metabolism. Moreover, spearman correlation analysis showed that the increased abundance of Escherichia-Shigella and decreased abundance of Clostridium_sensu_stricto_1 in IUGR piglets was closely associated with the alterations of slaughter weight, intestinal morphology, inflammatory cytokines, and plasma metabolites. Collectively, IUGR significantly impairs small intestine structure, modifies gut microbiota colonization, and disturbs inflammatory and metabolic profiles during the first 12 h after birth. The unbalanced gut microbiota mediated by IUGR contributes to the development of inflammation and metabolic diseases.
Collapse
Affiliation(s)
- Shimeng Huang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, P. R. China
| | - Na Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, P. R. China
| | - Cong Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Tiantian Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, P. R. China
| | - Wei Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, P. R. China
| | - Lili Jiang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Zhen Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China.,State Key Laboratory of Plant Physiology and Biochemistry, China Agricultural University, Beijing, 100193, P. R. China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, P. R. China
| | - Shiyu Tao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, P. R. China
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China. .,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, P. R. China.
| |
Collapse
|
33
|
Turkyilmaz IB, Arda Pirincci P, Bolkent S, Yanardag R. The effects of vitamins and selenium mixture or ranitidine against small intestinal injury induced by indomethacin in adult rats. J Food Biochem 2019; 43:e12808. [PMID: 31353593 DOI: 10.1111/jfbc.12808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/28/2018] [Revised: 01/03/2019] [Accepted: 01/31/2019] [Indexed: 01/18/2023]
Abstract
This study was aimed at investigating morphological and biochemical efficacies of antioxidants on indomethacin-induced small intestinal damage in rats. Group I: control animals (negative control) given only placebo, Group II: (positive control) are animals orally given combination of antioxidants [vitamin C (Vit C), vitamin E (Vit E), β-carotene and sodium selenite (Se)] daily for 3 days, Group III: Rats were given only indomethacin, Group IV: animals were given of antioxidants combination for 3 days, last dose was given 2 hr before the administration of indomethacin. Group V: Animals receiving ranitidine for 3 days (second positive control). Group VI: Animals received ranitidine for 3 days, last dose was given 2 hr before to indomethacin administration. Indomethacin caused degenerative morphological and biochemical changes, which were reversed on antioxidants administration. As a result, we propose that antioxidants combination would be therapeutically beneficial for treating indomethacin-induced lesions of small intestine. PRACTICAL APPLICATIONS: Indomethacin is a widely preferred nonsteroidal anti-inflammatory drug (NSAID) but its side effects on gastrointestinal system are well known. Indomethacin also causes production of reactive oxygen species. Antioxidants and selenium has protective effects. According to the results of this study, antioxidants and selenium can be used as a food supplement for preventing NSAID-induced side effects and toxicity.
Collapse
Affiliation(s)
- Ismet Burcu Turkyilmaz
- Faculty of Engineering, Department of Chemistry, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Pelin Arda Pirincci
- Faculty of Science, Department of Biology, Istanbul University, Istanbul, Turkey
| | - Sehnaz Bolkent
- Faculty of Science, Department of Biology, Istanbul University, Istanbul, Turkey
| | - Refiye Yanardag
- Faculty of Engineering, Department of Chemistry, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
34
|
Yang L, Tu D, Wang N, Deng Z, Zhan Y, Liu W, Hu Y, Liu T, Tan L, Li Y, Guo S, Wang A. The protective effects of DL-Selenomethionine against T-2/HT-2 toxins-induced cytotoxicity and oxidative stress in broiler hepatocytes. Toxicol In Vitro 2019; 54:137-146. [DOI: 10.1016/j.tiv.2018.09.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/13/2018] [Revised: 09/23/2018] [Accepted: 09/24/2018] [Indexed: 12/27/2022]
|
35
|
Impact of a dietary challenge with peroxidized oil on the glutathione redox status and integrity of the small intestine in weaned piglets. Animal 2018; 13:1641-1650. [PMID: 30458891 DOI: 10.1017/s1751731118003166] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/06/2022] Open
Abstract
Glutathione (GSH) is considered to play an important role in maintaining the integrity of the small intestine. In piglets, altered mucosal GSH levels might therefore be involved in weaning-induced changes of the small intestinal morphology and barrier function. To test this hypothesis, we aimed to challenge the mucosal GSH redox status during the first 28 days after weaning, by feeding diets containing 5% fresh linseed oil (CON), or 2.5% (OF1) or 5% (OF2) peroxidized linseed oil (peroxide value 225 mEq O2/kg oil) and exploring the effects on gut integrity. Piglets were pair-fed and had a total daily feed allowance of 32 g/kg BW. A fourth treatment included animals that were fed the control diet ad libitum (ACON). Animals were sampled at days 5 and 28 post-weaning. The malondialdehyde (MDA) concentration and GSH redox status (GSH/GSSG Eh) were determined in blood, liver and small intestinal mucosa. Histomorphology of the duodenal and jejunal mucosa was determined, and Ussing chambers were used to assess fluorescein isothiocyanate dextran (FD4) and horseradish peroxidase (HRP) fluxes across the mucosa. Results show that peroxidized linseed oil imposed an oxidative challenge at day 28, but not at day 5 post-weaning. At day 28, increasing levels of dietary peroxides to pair-fed pigs linearly increased MDA levels in duodenal and jejunal mucosa. Moreover, FD4 fluxes were significantly increased in OF1 (+75%) and OF2 (+64%) in the duodenum, and HRP fluxes tended (P=0.099) to show similar differences, as compared to CON. This co-occurred with a significant 11 mV increase of the hepatic GSH/GSSG Eh, potentiated by a significantly increased GSH peroxidase activity for treatments OF1 (+47%) and OF2 (+63%) in liver as compared to CON. Furthermore; duodenal HRP flux significantly correlated with the hepatic glutathione disulphide (GSSG) level (r=0.650), as also observed in the jejunum for hepatic GSSG (r=0.627) and GSH/GSSG Eh (r=0.547). The jejunal permeability was not affected, but FD4 and HRP fluxes significantly correlated with the local GSH (r=0.619; r=0.733) and GSSG (r=0.635; r=0586) levels. Small intestinal histomorphology was not affected by dietary lipid peroxides, nor were there any correlations found with the GSH redox system. To conclude, under oxidative stress conditions, jejunal barrier function is related to the local and hepatic GSH redox system. It is suggested that the hepatic GSH system participates in the elimination of luminal peroxides, and thereby impacts on duodenal barrier function.
Collapse
|
36
|
Cui K, Wang B, Zhang N, Tu Y, Ma T, Diao Q. iTRAQ-based quantitative proteomic analysis of alterations in the intestine of Hu sheep under weaning stress. PLoS One 2018; 13:e0200680. [PMID: 30024939 PMCID: PMC6053177 DOI: 10.1371/journal.pone.0200680] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/03/2017] [Accepted: 06/26/2018] [Indexed: 01/22/2023] Open
Abstract
When Lambs are weaned off ewe's milk, metabolic, structural, and functional changes often occur in the small intestine. Because information on the effects of weaning stress on the proteome of the intestine is limited, an animal model was established with eight pairs of twin lambs divided into artificially reared and ewe-reared groups, which was followed by proteome analysis using iTRAQ technology. Changes occurred in the morphology of the intestine and 5,338 proteins in three biological replicates with less than a 1.2% false discovery rate were identified and quantified. Among them, a subset of 389 proteins were screened as significantly up- (143) and down-regulated (246) in artificially reared compared with ewe-reared. According to Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis, the differentially expressed proteins that were strongly down-regulated were enriched in immune system processes, biological adhesion, and metabolic processes. The up-regulated proteins were enriched in gene expression, cellular biosynthetic processes, ribosome and RNA binding in response to weaning stress. A series of proteins associated with intestine morphology and immune function were identified, and levels of the mRNAs encoding these proteins were analyzed by real-time quantitative reverse transcription PCR. The results of this study increased our understanding of the response of lambs weaned off ewe's milk and helped to determine the mechanisms underlying weaning stress.
Collapse
Affiliation(s)
- Kai Cui
- Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Bo Wang
- Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Naifeng Zhang
- Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yan Tu
- Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Tao Ma
- Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qiyu Diao
- Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- * E-mail:
| |
Collapse
|
37
|
Moretti S, Mrakic-Sposta S, Roncoroni L, Vezzoli A, Dellanoce C, Monguzzi E, Branchi F, Ferretti F, Lombardo V, Doneda L, Scricciolo A, Elli L. Oxidative stress as a biomarker for monitoring treated celiac disease. Clin Transl Gastroenterol 2018; 9:157. [PMID: 29880904 PMCID: PMC5992147 DOI: 10.1038/s41424-018-0031-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/12/2017] [Revised: 04/11/2018] [Accepted: 05/07/2018] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION High levels of reactive oxygen species (ROS) and impaired antioxidant defense systems lead to oxidative stress (OxS) and tissue injury in different intestinal and extra intestinal conditions, including celiac disease (CD). The aim of the present study was to investigate the role and potential use of ROS and other biomarkers of OxS in the clinical management of CD. METHODS We collected duodenal specimens and blood samples from naïve patients (N-CD), patients on a gluten free diet (GFD) including responders (CD-GFD) and non-responders (NRCD). We measured plasmatic ROS production (electron paramagnetic resonance, EPR), lipid peroxidation (thiobarbituric acid-reactive substances, TBARS), protein oxidation (protein carbonyl, PC), total antioxidant capacity (TAC), nitric oxides and glutathione (GSH) in erythrocytes. RESULTS Fifty-four patients affected by CD were enrolled (17 N-CD, 18 CD-GFD and 19 NRCD; 44 F; age 44 ± 13 years). A significant increase of plasmatic OxS biomarkers (ROS, peroxidated lipids, oxidized proteins, and nitrate concentrations) and decrease of antioxidant species (TAC and GSH levels) were found in NRCD and N-CD compared to CD-GFD. Comparably, a significant direct relationship between the severity of duodenal atrophy, ROS production rates and TBARS was found; conversely, TAC and GSH presented an inverse correlation. DISCUSSION OxS is involved in CD tissue damage and correlates with the degree of duodenal atrophy. These findings suggest the possible role of OxS biomarkers as indicators of CD activity during the clinical follow-up.
Collapse
Affiliation(s)
- Sarah Moretti
- Institute of Bioimaging and Molecular Physiology, National Research Council (CNR), Via Fratelli Cervi 93, 20090, Segrate, Italy
| | - Simona Mrakic-Sposta
- Institute of Bioimaging and Molecular Physiology, National Research Council (CNR), Via Fratelli Cervi 93, 20090, Segrate, Italy
| | - Leda Roncoroni
- Center for Prevention and Diagnosis of Celiac Disease- Div. of Gastroenterology and Endoscopy, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milano, Italy
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Via Festa del Perdono, 20122, Milano, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Via Festa del Perdono, 20122, Milano, Italy
| | - Alessandra Vezzoli
- Institute of Bioimaging and Molecular Physiology, National Research Council (CNR), Via Fratelli Cervi 93, 20090, Segrate, Italy
| | - Cinzia Dellanoce
- Institute of Clinical Physiology, National Research Council (CNR), Niguarda Ca' Granda Hospital, Via G. Moruzzi 1, 56124, Pisa, Italy
| | - Erika Monguzzi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Via Festa del Perdono, 20122, Milano, Italy
| | - Federica Branchi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Via Festa del Perdono, 20122, Milano, Italy
| | - Francesca Ferretti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Via Festa del Perdono, 20122, Milano, Italy
| | - Vincenza Lombardo
- Center for Prevention and Diagnosis of Celiac Disease- Div. of Gastroenterology and Endoscopy, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milano, Italy
| | - Luisa Doneda
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Via Festa del Perdono, 20122, Milano, Italy
| | - Alice Scricciolo
- Center for Prevention and Diagnosis of Celiac Disease- Div. of Gastroenterology and Endoscopy, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milano, Italy
| | - Luca Elli
- Center for Prevention and Diagnosis of Celiac Disease- Div. of Gastroenterology and Endoscopy, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milano, Italy.
| |
Collapse
|
38
|
Wan J, Zhang J, Chen D, Yu B, Mao X, Zheng P, Yu J, Huang Z, Luo J, Luo Y, He J. Alginate oligosaccharide alleviates enterotoxigenicEscherichia coli-induced intestinal mucosal disruption in weaned pigs. Food Funct 2018; 9:6401-6413. [DOI: 10.1039/c8fo01551a] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022]
Abstract
Alginate oligosaccharide (AOS) is a non-toxic, non-immunogenic, non-carcinogenic and biodegradable product generated by depolymerisation of alginate, and exhibits various salutary properties.
Collapse
Affiliation(s)
- Jin Wan
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- People's Republic of China
| | - Jiao Zhang
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- People's Republic of China
| | - Daiwen Chen
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- People's Republic of China
| | - Bing Yu
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- People's Republic of China
| | - Xiangbing Mao
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- People's Republic of China
| | - Ping Zheng
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- People's Republic of China
| | - Jie Yu
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- People's Republic of China
| | - Zhiqing Huang
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- People's Republic of China
| | - Junqiu Luo
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- People's Republic of China
| | - Yuheng Luo
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- People's Republic of China
| | - Jun He
- Institute of Animal Nutrition
- Sichuan Agricultural University
- Chengdu 611130
- People's Republic of China
| |
Collapse
|
39
|
Zhou X, Zhang Y, He L, Wan D, Liu G, Wu X, Yin Y. Serine prevents LPS-induced intestinal inflammation and barrier damage via p53-dependent glutathione synthesis and AMPK activation. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.10.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/24/2022] Open
|
40
|
Uchida H, Nakajima Y, Ohtake K, Ito J, Morita M, Kamimura A, Kobayashi J. Protective effects of oral glutathione on fasting-induced intestinal atrophy through oxidative stress. World J Gastroenterol 2017; 23:6650-6664. [PMID: 29085210 PMCID: PMC5643286 DOI: 10.3748/wjg.v23.i36.6650] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 03/22/2017] [Revised: 07/14/2017] [Accepted: 07/24/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To determine whether oral glutathione (GSH) administration can alleviate the effects of fasting-induced intestinal atrophy in the small intestinal mucosa.
METHODS Rats were divided into eight groups. One group was fed ad libitum, another was fed ad libitum and received oral GSH, and six groups were administrated saline (SA) or GSH orally during fasting. Mucosal height, apoptosis, and cell proliferation in the jejunum were histologically evaluated. iNOS protein expression (by immunohistochemistry), nitrite levels (by high performance liquid chromatography, as a measure of NO production), 8-hydroxydeoxyguanosine formation (by ELISA, indicating ROS levels), glutathione/oxidized glutathione (GSH/GSSG) ratio (by enzymatic colorimetric detection), and γ-glutamyl transpeptidase (Ggt1) mRNA levels in the jejunum (by semi-quantitative RT-PCR) were also estimated.
RESULTS Oral GSH administration was demonstrated to drastically reduce fasting-induced intestinal atrophy in the jejunum. In particular, jejunal mucosal height was enhanced in GSH-treated animals compared to SA-treated animals [527.2 ± 6.9 for 50 mg/kg GSH, 567.6 ± 5.4 for 500 mg/kg GSH vs 483.1 ± 4.9 (μm), P < 0.01 at 72 h]. This effect was consistent with decreasing changes in GSH-treated animals compared to SA-treated animals for iNOS protein staining [0.337 ± 0.016 for 50 mg/kg GSH, 0.317 ± 0.017 for 500 mg/kg GSH vs 0.430 ± 0.023 (area of staining part/area of tissue), P < 0.01 at 72 h] and NO [2.99 ± 0.29 for 50 mg/kg GSH, 2.88 ± 0.19 for 500 mg/kg GSH vs 5.34 ± 0.35 (nmol/g tissue), P < 0.01 at 72 h] and ROS [3.92 ± 0.46 for 50 mg/kg GSH, 4.58 ± 0.29 for 500 mg/kg GSH vs 6.42 ± 0.52 (8-OHdG pg/μg DNA), P < 0.01, P < 0.05 at 72 h, respectively] levels as apoptosis mediators in the jejunum. Furthermore, oral GSH administration attenuated cell proliferation decreases in the fasting jejunum [182.5 ± 1.9 for 500 mg/kg GSH vs 155.8 ± 3.4 (5-BrdU positive cells/10 crypts), P < 0.01 at 72 h]. Notably, both GSH concentration and Ggt1 mRNA expression in the jejunum were also attenuated in rats following oral administration of GSH during fasting as compared with fasting alone [0.45 ± 0.12 vs 0.97 ± 0.06 (nmol/mg tissue), P < 0.01; 1.01 ± 0.11 vs 2.79 ± 0.39 (Ggt1 mRNA/Gapdh mRNA), P < 0.01 for 500 mg/kg GSH at 48 h, respectively].
CONCLUSION Oral GSH administration during fasting enhances jejunal regenerative potential to minimize intestinal mucosal atrophy by diminishing fasting-mediated ROS generation and enterocyte apoptosis and enhancing cell proliferation.
Collapse
Affiliation(s)
- Hiroyuki Uchida
- Division of Pathophysiology, Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmaceutical Science, Josai University, Sakado, Saitama 350-0295, Japan
| | - Yukari Nakajima
- Division of Pathophysiology, Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmaceutical Science, Josai University, Sakado, Saitama 350-0295, Japan
| | - Kazuo Ohtake
- Division of Pathophysiology, Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmaceutical Science, Josai University, Sakado, Saitama 350-0295, Japan
| | - Junta Ito
- Division of Pathophysiology, Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmaceutical Science, Josai University, Sakado, Saitama 350-0295, Japan
| | - Masahiko Morita
- Kyowa Hakko Bio Co., Ltd. Healthcare Products Development Center, Tsukuba-shi, Ibaraki 305-0841, Japan
| | - Ayako Kamimura
- Kyowa Hakko Bio Co., Ltd. Healthcare Products Development Center, Tsukuba-shi, Ibaraki 305-0841, Japan
| | - Jun Kobayashi
- Division of Pathophysiology, Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmaceutical Science, Josai University, Sakado, Saitama 350-0295, Japan
| |
Collapse
|
41
|
|
42
|
Kumar S, Lal S, Bhatnagar A. Regulatory T cell subsets in peripheral blood of celiac disease patients and TLR2 expression: correlation with oxidative stress. APMIS 2017; 125:888-901. [DOI: 10.1111/apm.12735] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/28/2017] [Accepted: 05/22/2017] [Indexed: 01/21/2023]
Affiliation(s)
- Sanjay Kumar
- Department of Biochemistry; Panjab University; Chandigarh India
| | - Sadhna Lal
- Department of Gastroenterology; Postgraduate Institute of Medical Education & Research; Chandigarh India
| | | |
Collapse
|
43
|
Vergauwen H, Degroote J, Prims S, Wang W, Fransen E, De Smet S, Casteleyn C, Van Cruchten S, Michiels J, Van Ginneken C. Artificial rearing influences the morphology, permeability and redox state of the gastrointestinal tract of low and normal birth weight piglets. J Anim Sci Biotechnol 2017; 8:30. [PMID: 28405313 PMCID: PMC5385054 DOI: 10.1186/s40104-017-0159-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/05/2016] [Accepted: 03/16/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND In this study the physiological implications of artificial rearing were investigated. Low (LBW) and normal birth weight (NBW) piglets were compared as they might react differently to stressors caused by artificial rearing. In total, 42 pairs of LBW and NBW piglets from 16 litters suckled the sow until d19 of age or were artificially reared starting at d3 until d19 of age. Blood and tissue samples that were collected after euthanasia at 0, 3, 5, 8 and 19 d of age. Histology, ELISA, and Ussing chamber analysis were used to study proximal and distal small intestine histo-morphology, proliferation, apoptosis, tight junction protein expression, and permeability. Furthermore, small intestine, liver and systemic redox parameters (GSH, GSSG, GSH-Px and MDA) were investigated using HPLC. RESULTS LBW and NBW artificially reared piglets weighed respectively 40 and 33% more than LBW and NBW sow-reared piglets at d19 (P < 0.01). Transferring piglets to a nursery at d3 resulted in villus atrophy, increased intestinal FD-4 and HRP permeability and elevated GSSG/GSH ratio in the distal small intestine at d5 (P < 0.05). GSH concentrations in the proximal small intestine remained stable, while they decreased in the liver (P < 0.05). From d5 until d19, villus width and crypt depth increased, whereas PCNA, caspase-3, occludin and claudin-3 protein expressions were reduced. GSH, GSSG and permeability recovered in artificially reared piglets (P < 0.05). CONCLUSION The results suggest that artificial rearing altered the morphology, permeability and redox state without compromising piglet performance. The observed effects were not depending on birth weight.
Collapse
Affiliation(s)
- Hans Vergauwen
- Laboratory of Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Biomedical, Pharmaceutical and Veterinary Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, D.U.015, 2610 Wilrijk, Belgium
| | - Jeroen Degroote
- Department of Applied Biosciences, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Sara Prims
- Laboratory of Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Biomedical, Pharmaceutical and Veterinary Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, D.U.015, 2610 Wilrijk, Belgium
| | - Wei Wang
- Department of Applied Biosciences, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium.,Laboratory for Animal Nutrition and Animal Product Quality (LANUPRO), Department of Animal Production, Faculty of Bioscience Engineering, Ghent University, Melle, Belgium
| | - Erik Fransen
- StatUa Center for Statistics, University of Antwerp, Antwerp, Belgium
| | - Stefaan De Smet
- Laboratory for Animal Nutrition and Animal Product Quality (LANUPRO), Department of Animal Production, Faculty of Bioscience Engineering, Ghent University, Melle, Belgium
| | - Christophe Casteleyn
- Laboratory of Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Biomedical, Pharmaceutical and Veterinary Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, D.U.015, 2610 Wilrijk, Belgium
| | - Steven Van Cruchten
- Laboratory of Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Biomedical, Pharmaceutical and Veterinary Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, D.U.015, 2610 Wilrijk, Belgium
| | - Joris Michiels
- Department of Applied Biosciences, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Chris Van Ginneken
- Laboratory of Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Biomedical, Pharmaceutical and Veterinary Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, D.U.015, 2610 Wilrijk, Belgium
| |
Collapse
|
44
|
Li J, Liu Y, Kim E, March JC, Bentley WE, Payne GF. Electrochemical reverse engineering: A systems-level tool to probe the redox-based molecular communication of biology. Free Radic Biol Med 2017; 105:110-131. [PMID: 28040473 DOI: 10.1016/j.freeradbiomed.2016.12.029] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 10/07/2016] [Revised: 12/06/2016] [Accepted: 12/20/2016] [Indexed: 12/20/2022]
Abstract
The intestine is the site of digestion and forms a critical interface between the host and the outside world. This interface is composed of host epithelium and a complex microbiota which is "connected" through an extensive web of chemical and biological interactions that determine the balance between health and disease for the host. This biology and the associated chemical dialogues occur within a context of a steep oxygen gradient that provides the driving force for a variety of reduction and oxidation (redox) reactions. While some redox couples (e.g., catecholics) can spontaneously exchange electrons, many others are kinetically "insulated" (e.g., biothiols) allowing the biology to set and control their redox states far from equilibrium. It is well known that within cells, such non-equilibrated redox couples are poised to transfer electrons to perform reactions essential to immune defense (e.g., transfer from NADH to O2 for reactive oxygen species, ROS, generation) and protection from such oxidative stresses (e.g., glutathione-based reduction of ROS). More recently, it has been recognized that some of these redox-active species (e.g., H2O2) cross membranes and diffuse into the extracellular environment including lumen to transmit redox information that is received by atomically-specific receptors (e.g., cysteine-based sulfur switches) that regulate biological functions. Thus, redox has emerged as an important modality in the chemical signaling that occurs in the intestine and there have been emerging efforts to develop the experimental tools needed to probe this modality. We suggest that electrochemistry provides a unique tool to experimentally probe redox interactions at a systems level. Importantly, electrochemistry offers the potential to enlist the extensive theories established in signal processing in an effort to "reverse engineer" the molecular communication occurring in this complex biological system. Here, we review our efforts to develop this electrochemical tool for in vitro redox-probing.
Collapse
Affiliation(s)
- Jinyang Li
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA; Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD, USA
| | - Yi Liu
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA; Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD, USA
| | - Eunkyoung Kim
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA; Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD, USA
| | - John C March
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - William E Bentley
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA; Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD, USA
| | - Gregory F Payne
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA; Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD, USA.
| |
Collapse
|
45
|
Eltahawy NA, Elsonbaty SM, Abunour S, Zahran WE. Synergistic effect of aluminum and ionizing radiation upon ultrastructure, oxidative stress and apoptotic alterations in Paneth cells of rat intestine. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2017; 24:6657-6666. [PMID: 28083743 DOI: 10.1007/s11356-017-8392-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 04/24/2016] [Accepted: 01/04/2017] [Indexed: 06/06/2023]
Abstract
Environmental and occupational exposure to aluminum along with ionizing radiation results in serious health problems. This study was planned to investigate the impact of oxidative stress provoked by exposure to ionizing radiation with aluminum administration upon cellular ultra structure and apoptotic changes in Paneth cells of rat small intestine . Animals received daily aluminum chloride by gastric gavage at a dose 0.5 mg/Kg BW for 4 weeks. Whole body gamma irradiation was applied at a dose 2 Gy/week up to 8 Gy. Ileum malondialdehyde, advanced oxidative protein products, protein carbonyl and tumor necrosis factor-alpha were assessed as biomarkers of lipid peroxidation, protein oxidation and inflammation respectively along with superoxide dismutase, catalase, and glutathione peroxidase activities as enzymatic antioxidants. Moreover, analyses of cell cycle division and apoptotic changes were evaluated by flow cytometry. Intestinal cellular ultra structure was investigated using transmission electron microscope.Oxidative and inflammatory stresses assessment in the ileum of rats revealed that aluminum and ionizing radiation exposures exhibited a significant effect upon the increase in oxidative stress biomarkers along with the inflammatory marker tumor necrosis factor-α accompanied by a significant decreases in the antioxidant enzyme activities. Flow cytometric analyses showed significant alterations in the percentage of cells during cell cycle division phases along with significant increase in apoptotic cells. Ultra structurally, intestinal cellular alterations with marked injury in Paneth cells at the sites of bacterial translocation in the crypt of lumens were recorded. The results of this study have clearly showed that aluminum and ionizing radiation exposures induced apoptosis with oxidative and inflammatory disturbance in the Paneth cells of rat intestine, which appeared to play a major role in the pathogenesis of cellular damage. Furthermore, the interaction of these two intestinal toxic routes was found to be synergistic.
Collapse
Affiliation(s)
- N A Eltahawy
- National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - S M Elsonbaty
- National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - S Abunour
- National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - W E Zahran
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
46
|
Pérez S, Taléns-Visconti R, Rius-Pérez S, Finamor I, Sastre J. Redox signaling in the gastrointestinal tract. Free Radic Biol Med 2017; 104:75-103. [PMID: 28062361 DOI: 10.1016/j.freeradbiomed.2016.12.048] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 07/02/2016] [Revised: 12/20/2016] [Accepted: 12/31/2016] [Indexed: 12/16/2022]
Abstract
Redox signaling regulates physiological self-renewal, proliferation, migration and differentiation in gastrointestinal epithelium by modulating Wnt/β-catenin and Notch signaling pathways mainly through NADPH oxidases (NOXs). In the intestine, intracellular and extracellular thiol redox status modulates the proliferative potential of epithelial cells. Furthermore, commensal bacteria contribute to intestine epithelial homeostasis through NOX1- and dual oxidase 2-derived reactive oxygen species (ROS). The loss of redox homeostasis is involved in the pathogenesis and development of a wide diversity of gastrointestinal disorders, such as Barrett's esophagus, esophageal adenocarcinoma, peptic ulcer, gastric cancer, ischemic intestinal injury, celiac disease, inflammatory bowel disease and colorectal cancer. The overproduction of superoxide anion together with inactivation of superoxide dismutase are involved in the pathogenesis of Barrett's esophagus and its transformation to adenocarcinoma. In Helicobacter pylori-induced peptic ulcer, oxidative stress derived from the leukocyte infiltrate and NOX1 aggravates mucosal damage, especially in HspB+ strains that downregulate Nrf2. In celiac disease, oxidative stress mediates most of the cytotoxic effects induced by gluten peptides and increases transglutaminase levels, whereas nitrosative stress contributes to the impairment of tight junctions. Progression of inflammatory bowel disease relies on the balance between pro-inflammatory redox-sensitive pathways, such as NLRP3 inflammasome and NF-κB, and the adaptive up-regulation of Mn superoxide dismutase and glutathione peroxidase 2. In colorectal cancer, redox signaling exhibits two Janus faces: On the one hand, NOX1 up-regulation and derived hydrogen peroxide enhance Wnt/β-catenin and Notch proliferating pathways; on the other hand, ROS may disrupt tumor progression through different pro-apoptotic mechanisms. In conclusion, redox signaling plays a critical role in the physiology and pathophysiology of gastrointestinal tract.
Collapse
Affiliation(s)
- Salvador Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Burjasot, 46100 Valencia, Spain
| | - Raquel Taléns-Visconti
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Burjasot, 46100 Valencia, Spain
| | - Sergio Rius-Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Burjasot, 46100 Valencia, Spain
| | - Isabela Finamor
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Burjasot, 46100 Valencia, Spain
| | - Juan Sastre
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Burjasot, 46100 Valencia, Spain.
| |
Collapse
|
47
|
Mandal P. Potential biomarkers associated with oxidative stress for risk assessment of colorectal cancer. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:557-565. [PMID: 28229171 DOI: 10.1007/s00210-017-1352-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/06/2016] [Accepted: 01/30/2017] [Indexed: 02/07/2023]
Abstract
Cells are continuously threatened by the damage caused by reactive oxygen/nitrogen species (ROS/RNS), which are produced during physiological oxygen metabolism. In our review, we will summarize the latest reports on the role of oxidative stress and oxidative stress-induced signaling pathways in the etiology of colorectal cancer. The differences in ROS generation may influence the levels of oxidized proteins, lipids, and DNA damage, thus contributing to the higher susceptibility of colon. Reactive species (RS) of various types are formed and are powerful oxidizing agents, capable of damaging DNA and other biomolecules. Increased formation of RS can promote the development of malignancy, and the "normal" rates of RS generation may account for the increased risk of cancer development in the aged. In this review, we focus on the role of oxidative stress in the etiology of colorec-tal cancer and discuss free radicals and free radical-stimulated pathways in colorectal carcinogenesis.
Collapse
Affiliation(s)
- Paramita Mandal
- Department of Zoology, The University of Burdwan, Burdwan, 713104, West Bengal, India.
| |
Collapse
|
48
|
Demethyleneberberine alleviates inflammatory bowel disease in mice through regulating NF-κB signaling and T-helper cell homeostasis. Inflamm Res 2016; 66:187-196. [PMID: 27900412 DOI: 10.1007/s00011-016-1005-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/18/2016] [Revised: 10/24/2016] [Accepted: 10/28/2016] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE The activation of NF-κB signaling and unbalance of T-helper (Th) cells have been reported to play a key role in the pathogenesis of colitis. Cortex Phellodendri Chinensis (CPC) is commonly used to treat inflammation and diarrhea. Demethyleneberberine (DMB), a component of CPC, was reported to treat alcoholic liver disease as a novel natural mitochondria-targeted antioxidant in our previous study. In this study, we investigated whether DMB could protect against dextran sulfate sodium (DSS)-induced inflammatory colitis in mice by regulation of NF-κB pathway and Th cells homeostatis. METHODS Inflammatory colitis mice were induced by 3% DSS, and DMB were orally administered on the doses of 150 and 300 mg/kg. In vitro, DMB (10, 20, 40 μM) and N-acetyl cysteine (NAC, 5 mM) were co-cultured with RAW264.7 for 2 h prior to lipopolysaccharide (LPS) stimulation, and splenocytes from the mice were cultured ex vivo for 48 h for immune response test. RESULTS In vivo, DMB significantly alleviated the weight loss and diminished myeloperoxidase (MPO) activity, while significantly reduced the production of pro-inflammatory cytokines, such as interleukin (IL)-6 and tumor necrosis factor-α (TNF-α), and inhibited the activation of NF-κB signaling pathway. Furthermore, DMB decreased interferon (IFN)-γ, increased IL-4 concentration in the mice splenocytes and the ratio of IgG1/IgG2a in the serum. In vitro, ROS production and pro-inflammation cytokines were markedly inhibited by DMB in RAW264.7 cell. CONCLUSIONS Our findings revealed that DMB alleviated mice colitis and inhibited the inflammatory responses by inhibiting NF-κB pathway and regulating the balance of Th cells.
Collapse
|
49
|
Vergauwen H, Prims S, Degroote J, Wang W, Casteleyn C, van Cruchten S, de Smet S, Michiels J, van Ginneken C. In Vitro Investigation of Six Antioxidants for Pig Diets. Antioxidants (Basel) 2016; 5:antiox5040041. [PMID: 27845706 PMCID: PMC5187539 DOI: 10.3390/antiox5040041] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/07/2016] [Revised: 10/03/2016] [Accepted: 11/02/2016] [Indexed: 12/26/2022] Open
Abstract
Oxidative stress in the small intestinal epithelium can lead to barrier malfunction. In this study, the effect of rosmarinic acid (RA), quercetin (Que), gallic acid (GA), lipoic acid (LA), ethoxyquin (ETQ) and Se-methionine (SeMet) pre-treatments using 2 mM Trolox as a control on the viability and the generation of intracellular reactive oxygen species (iROS) of oxidatively (H₂O₂) stressed intestinal porcine epithelial cells (IPEC-J2) was investigated. A neutral red assay showed that RA (50-400 µM), Que (12.5-200 µM), GA (50-400 µM), ETQ (6.25-100 µM), and SeMet (125-1000 µM) pre-treatments but not LA significantly increased the viability of H₂O₂-stressed IPEC-J2 cells (p < 0.05). A 5-(and-6)-chloromethyl-2',7'-dichlorodihydrofluorescein diacetate, acetyl ester (CM-H₂DCFDA) fluorescent probe showed that RA (100-600 µM), Que (25-800 µM), ETQ (3.125-100 µM) and SeMet (500-2000 µM) pre-treatments significantly reduced iROS in IPEC-J2 monolayers (p < 0.05). Moreover, RA and Que were most effective in reducing iROS. Therefore, the effects of RA and Que on barrier functioning in vitro were examined. RA and Que pre-treatments significantly decreased fluorescein isothiocyanate (FITC)-conjugated dextran-4 (4 kDa) permeability and transepithelial electrical resistance (TEER) of an IPEC-J2 cell monolayer (p < 0.05). These in vitro results of RA and Que hold promise for their use as antioxidants in pig feed.
Collapse
Affiliation(s)
- Hans Vergauwen
- Laboratory of Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Biomedical, University of Antwerp, Wilrijk 2610, Belgium.
| | - Sara Prims
- Laboratory of Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Biomedical, University of Antwerp, Wilrijk 2610, Belgium.
| | - Jeroen Degroote
- Department of Applied Biosciences, Faculty of Bioscience Engineering, Ghent University, Ghent 9000, Belgium.
| | - Wei Wang
- Department of Applied Biosciences, Faculty of Bioscience Engineering, Ghent University, Ghent 9000, Belgium.
- Laboratory for Animal Nutrition and Animal Product Quality (LANUPRO), Department of Animal Production, Faculty of Bioscience Engineering, Ghent University, Melle 9090, Belgium.
| | - Christophe Casteleyn
- Laboratory of Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Biomedical, University of Antwerp, Wilrijk 2610, Belgium.
| | - Steven van Cruchten
- Laboratory of Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Biomedical, University of Antwerp, Wilrijk 2610, Belgium.
| | - Stefaan de Smet
- Laboratory for Animal Nutrition and Animal Product Quality (LANUPRO), Department of Animal Production, Faculty of Bioscience Engineering, Ghent University, Melle 9090, Belgium.
| | - Joris Michiels
- Department of Applied Biosciences, Faculty of Bioscience Engineering, Ghent University, Ghent 9000, Belgium.
| | - Chris van Ginneken
- Laboratory of Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Biomedical, University of Antwerp, Wilrijk 2610, Belgium.
| |
Collapse
|
50
|
Liu Z, Qu Y, Wang J, Wu R. Selenium Deficiency Attenuates Chicken Duodenal Mucosal Immunity via Activation of the NF-κb Signaling Pathway. Biol Trace Elem Res 2016; 172:465-473. [PMID: 26728795 DOI: 10.1007/s12011-015-0589-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 09/25/2015] [Accepted: 12/11/2015] [Indexed: 02/07/2023]
Abstract
Selenium (Se) deficiency can cause intestinal mucosal inflammation, which is related to activation of nuclear transcription factor kappa-B (NF-κB) signaling pathway. However, the mechanism of inflammatory response in chicken duodenal mucosa caused by Se deficiency and its relationship with the NF-κB signaling pathway remain elusive. In this study, we firstly obtained Se-deficient chickens bred with 0.01 mg/kg Se and the normal chickens bred with 0.4 mg/kg Se for 35 days. Then, NF-κB signaling pathway, secretory immunoglobulin A (SIgA), inflammatory cytokines, oxidized glutathione, glutathione peroxidase, and glutathione activities were determined. The results showed that Se deficiency obviously enhanced p50, p65, and p65 DNA-binding activities. The phosphorylation of IκB-α and phosphorylation of kappa-B kinase subunit alpha (IKKα) and IKKα were elevated, but IκB-α was decreased (P < 0.05). Moreover, Se deficiency reduced SIgA amount in the duodenal mucosa but increased the level of interleukin-1β (IL-1β), IL-17A, tumor necrosis factor-α (TNF-α), and interferon gamma (IFN-γ). In contrast, anti-inflammatory cytokines, such as TGF-β1 and IL-10, were significantly suppressed. Additionally, Se deficiency increased oxidized glutathione activity, whereas decreased glutathione peroxidase and glutathione activities (P < 0.05), suggesting that Se deficiency affected the regulation function of redox. Taken together, our results demonstrated that Se deficiency attenuated chicken duodenal mucosal immunity via activation of NF-κB signaling pathway regulated by redox activity, which suggested that Se is a crucial host factor involved in regulating inflammation.
Collapse
Affiliation(s)
- Zhe Liu
- College of Life Sciences and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, People's Republic of China
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, 2 Xinyang Road, Daqing, Heilongjiang, 163319, People's Republic of China
| | - Yanpeng Qu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, 2 Xinyang Road, Daqing, Heilongjiang, 163319, People's Republic of China
| | - Jianfa Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, 2 Xinyang Road, Daqing, Heilongjiang, 163319, People's Republic of China
| | - Rui Wu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, 2 Xinyang Road, Daqing, Heilongjiang, 163319, People's Republic of China.
| |
Collapse
|