1
|
Chen J, Lei KW, Li SY, Li DP, Wang YL, Wang X, Bai X, Huang YL. Dose effects of iron on growth, antioxidant potential, intestinal morphology, and intestinal barrier in yellow-feathered broilers. Poult Sci 2025; 104:104865. [PMID: 39919560 PMCID: PMC11851222 DOI: 10.1016/j.psj.2025.104865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
This experiment was conducted to investigate the dose effects of iron on growth performance, antioxidant function, small intestinal histology, and intestinal barrier of 63-day-old yellow-feathered broilers. A total of 720 1-day-old male yellow-feathered broilers were randomly divided into 9 treatments, with 8 cages per treatment and 10 birds per cage. The Fe supplementation was 0, 20, 40, 60, 80, 160, 320, 640, and 1280 mg/kg, respectively, in the form of FeSO4•7H2O. The results showed that the ADG (P = 0.002) and ADFI (P < 0.001) decreased linearly with increased dietary Fe supplementation. Malondialdehyde (MDA) concentration in plasma (P = 0.001), duodenum (P < 0.001), and jejunum (P < 0.001) were increased linearly as dietary Fe increased. As dietary Fe increased, there was a linear decrease in the villus height and the villus height/crypt depth in the duodenum (P = 0.003; P = 0.001) and jejunum (P = 0.001; P < 0.001). Decreased secretory immunoglobulin A (sIgA) concentration in jejunal mucosa (P < 0.001) was observed with increased dietary Fe concentration. Lower jejunal sIgA concentrations were observed in birds consuming more than 160 mg/kg of Fe (P < 0.001). A quadratic response was found for jejunal diamine oxidase (DAO) activity (P = 0.011) as dietary Fe supplementation was increased. The highest response of DAO in jejunal mucosa was observed for broilers supplemented with 160 mg/kg of Fe. Furthermore, the mRNA expressions of ZO-1 (P < 0.001), occludin (P = 0.004), and claudin-1 (P = 0.007) in jejunal mucosa decreased linearly with increased dietary Fe concentration. Data from the study suggests that there is no need to supplement additional Fe to a corn-soybean-based diet for yellow-feathered broilers based on growth performance, antioxidant potential, small intestinal histology, and intestinal barrier. Chronic iron exposure (≥ 160 mg/kg) can damage the intestinal barrier function, and further increase of Fe supplementation can lead to oxidative stress and even cause growth inhibition for yellow-feathered broilers.
Collapse
Affiliation(s)
- J Chen
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - K W Lei
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - S Y Li
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - D P Li
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - Y L Wang
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - X Wang
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - X Bai
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - Y L Huang
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China.
| |
Collapse
|
2
|
Johnson AJ, Li W, Dittrich BI, Cole AC, Prodell MK, Lyons JW, Fritz SA, Fregulia P, Chen C, Kwon CH, Jang YD. Effect of second iron injection on growth performance, hematological parameters, and fecal microbiome of piglets fed different dietary iron levels. J Anim Sci 2025; 103:skae371. [PMID: 39657578 DOI: 10.1093/jas/skae371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024] Open
Abstract
This experiment was conducted to evaluate the effects of a second iron injection for suckling pigs fed diets with different dietary iron levels in the nursery period on growth performance, hematological parameters, serum and liver trace mineral content, fecal score, microbiome, and metabolites. A total of 70 newborn pigs from 7 litters were assigned to either 1 or 2 iron injections within the litter and received the first i.m. iron injection (200 mg) at 2 to 3 d of age. Pigs assigned to the second injection treatment received an additional iron injection 5 d after the first injection. At weaning (days 27 to 30 of age), pigs within iron injection treatments were divided into 2 nursery diet treatments for a 27-d growth period. Treatments were 1) no additional iron injection + nursery diets with 100 ppm iron (NC), 2) second i.m. iron injection (200 mg) + NC diets, 3) no additional iron injection + nursery diets with 200 ppm iron (PC), and 4) second i.m. iron injection (200 mg) + PC diets. The second iron injection increased liver iron content at weaning (P = 0.08, tendency), and serum iron, hemoglobin, and hematocrit levels until day 13 postweaning (P < 0.05). In the nursery period, pigs receiving the second iron injection had a greater final body weight (P = 0.08, tendency), overall growth rate (P = 0.08, tendency) and feed intake (P < 0.05), and lower fecal score (P < 0.05) indicating firmer feces compared to those receiving 1 iron injection. There was no major effect of dietary iron level or interaction with the iron injection treatment in any measurements except that the pigs fed the PC diets had greater hemoglobin and hematocrit levels (P < 0.05) at day 27 postweaning and a lower fecal score (P = 0.08, tendency) in the late nursery period than those fed the NC diets. The second iron injection reduced fecal bacterial alpha-diversity based on Faith's phylogenetic diversity at weaning (P < 0.05), while the second iron injection and dietary iron levels resulted in dissimilarity in the fecal bacterial community based on Unweighted Unifrac analysis (P < 0.05; at weaning by iron injection and day 27 postweaning by dietary iron level). In conclusion, the second iron injection for suckling pigs improved postweaning growth performance and hemoglobin levels and affected the fecal microbiome, whereas an additional 100 ppm of dietary iron supplementation increased hemoglobin levels and altered the fecal microbiome in the late nursery period but did not affect postweaning growth.
Collapse
Affiliation(s)
- Alaina J Johnson
- Department of Animal and Food Science, University of Wisconsin-River Falls, River Falls, WI 54022, USA
| | - Wenli Li
- United States Department of Agriculture - Agricultural Research Service, US Dairy Forage Research Center, Madison, WI, 53706, USA
| | - Barbara I Dittrich
- Department of Animal and Food Science, University of Wisconsin-River Falls, River Falls, WI 54022, USA
| | - Aleah C Cole
- Department of Animal and Food Science, University of Wisconsin-River Falls, River Falls, WI 54022, USA
| | - Marie K Prodell
- Department of Animal and Food Science, University of Wisconsin-River Falls, River Falls, WI 54022, USA
| | | | - Scott A Fritz
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS 66506, USA
| | - Priscila Fregulia
- United States Department of Agriculture - Agricultural Research Service, US Dairy Forage Research Center, Madison, WI, 53706, USA
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, USA
| | - Chi Chen
- Department of Animal Science, University of Minnesota, St. Paul, MN 55108, USA
| | - Chan Ho Kwon
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA
| | - Young Dal Jang
- Department of Animal and Food Science, University of Wisconsin-River Falls, River Falls, WI 54022, USA
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
3
|
Deng S, Yang W, Fang C, He H, Liu J, Fang R. New insights into the mechanisms of iron absorption: Iron dextran uptake in the intestines of weaned pigs through glucose transporter 5 (GLUT5) and divalent metal transporter 1 (DMT1) transporters. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 19:25-40. [PMID: 39628646 PMCID: PMC11612655 DOI: 10.1016/j.aninu.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/17/2024] [Accepted: 05/03/2024] [Indexed: 12/06/2024]
Abstract
The purpose of this study was to gain insight into the mechanism of iron dextran (DexFe) absorption in the intestines. A total of 72 piglets (average BW = 7.12 ± 0.75 kg, male to female ratio = 1:1) weaned at 28 d of age were randomly divided into two treatment groups with six replicates for each group. The experimental diets included the basal diet supplemented with 100 mg/kg iron dextran (DexFe group) and the basal diet supplemented with 100 mg/kg FeSO4·H2O (CON group). The experiment lasted for 28 d. The piglets' intestinal iron transport was measured in vitro using an Ussing chamber. Porcine intestinal epithelial cell line (IPEC-J2) cells were used to develop a monolayer cell model that explored the molecular mechanism of DexFe absorption. Results showed that compared to the CON group, the ADG of pigs in the DexFe group was improved (P = 0.022), while the F/G was decreased (P = 0.015). The serum iron concentration, apparent iron digestibility, and iron deposition in the duodenum, jejunum, and ileum were increased (P < 0.05) by dietary DexFe supplementation. Piglets in the DexFe group had higher serum red blood count, hemoglobin, serum iron content, serum ferritin and transferrin levels and lower total iron binding capacity (P < 0.05). In the Ussing chamber test, the iron absorption rate of the DexFe group was greater (P < 0.001) than the CON group, and there was no significant difference between the DexFe group and the glucose group (P > 0.05). Furthermore, when compared to the CON group, DexFe administration improved (P < 0.05) SLC2A5 gene and glucose transporter 5 (GLUT5) protein expression but had no effect (P > 0.05) on SLC11A2 gene or divalent metal transporter 1 (DMT1) protein expression. Once the GLUT5 protein was suppressed, the iron transport rate and apparent permeability coefficient were decreased (P < 0.05) in IPEC-J2 monolayer cell models. The findings suggest the effectiveness of DexFe application in weaned piglets and revealed for the first time that DexFe absorption in the intestine is closely related to the glucose transporter GLUT5 protein channel.
Collapse
Affiliation(s)
- Shengting Deng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Research Center of Intelligent Animal Husbandry, Changsha 410128, China
| | - Weiguang Yang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Research Center of Intelligent Animal Husbandry, Changsha 410128, China
| | - Chengkun Fang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Haosheng He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Research Center of Intelligent Animal Husbandry, Changsha 410128, China
| | - Jiamin Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Research Center of Intelligent Animal Husbandry, Changsha 410128, China
| | - Rejun Fang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Research Center of Intelligent Animal Husbandry, Changsha 410128, China
| |
Collapse
|
4
|
Soriano-Lerma A, García-Burgos M, Barton W, Alférez MJM, Crespo-Pérez JV, Soriano M, López-Aliaga I, Cotter PD, García-Salcedo JA. Comprehensive insight into the alterations in the gut microbiome and the intestinal barrier as a consequence of iron deficiency anaemia. Biomed J 2024; 47:100701. [PMID: 38281699 PMCID: PMC11550200 DOI: 10.1016/j.bj.2024.100701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 11/09/2023] [Accepted: 01/19/2024] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Iron deficiency is the top leading cause of anaemia, whose treatment has been shown to deteriorate gut health. However, a comprehensive analysis of the intestinal barrier and the gut microbiome during iron deficiency anemia (IDA) has not been performed to date. This study aims to delve further into the analysis of these two aspects, which will mean a step forward minimising the negative impact of iron supplements on intestinal health. METHODS IDA was experimentally induced in an animal model. Shotgun sequencing was used to analyse the gut microbiome in the colonic region, while the intestinal barrier was studied through histological analyses, mRNA sequencing (RNA-Seq), qPCR and immunofluorescence assays. Determinations of lipopolysaccharide (LPS) and bacteria-specific immunoglobulins were performed to assess microbial translocation. RESULTS Microbial metabolism in the colon shifted towards an increased production of certain amino acids, short chain fatty acids and nucleotides, with Clostridium species being enriched during IDA. Structural alterations of the colonic epithelium were shown by histological analysis. RNA-Seq revealed a downregulation of extracellular matrix-associated genes and proteins and an overall underdeveloped epithelium. Increased levels of serum LPS and an increased immune response against dysbiotic bacteria support an impairment in the integrity of the gut barrier during IDA. CONCLUSIONS IDA negatively impacts the gut microbiome and the intestinal barrier, triggering an increased microbial translocation. This study emphasizes the deterioration of gut health during IDA and the fact that it should be addressed when treating the disease.
Collapse
Affiliation(s)
- Ana Soriano-Lerma
- Department of Physiology (Faculty of Pharmacy, Campus Universitario de Cartuja), Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, Granada, Spain; GENYO, Centre for Genomics and Oncological Research: Pfizer / University of Granada / Andalusian Regional Government, Granada, Spain; Instituto de Investigación Biosanitaria ibs, Granada, Spain
| | - María García-Burgos
- Department of Physiology (Faculty of Pharmacy, Campus Universitario de Cartuja), Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, Granada, Spain; GENYO, Centre for Genomics and Oncological Research: Pfizer / University of Granada / Andalusian Regional Government, Granada, Spain
| | - Wiley Barton
- VistaMilk SFI Research Centre, Cork, Ireland; Teagasc Food Research Centre, Carlow, Ireland
| | - María José M Alférez
- Department of Physiology (Faculty of Pharmacy, Campus Universitario de Cartuja), Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, Granada, Spain
| | - Jorge Valentín Crespo-Pérez
- Service of Anatomical pathology, Intercenter Regional Unit Granada, University Hospital Virgen de las Nieves, Granada, Spain
| | - Miguel Soriano
- Center for Intensive Mediterranean Agrosystems and Agri-food Biotechnology (CIAIMBITAL), University of Almeria, Almeria, Spain.
| | - Inmaculada López-Aliaga
- Department of Physiology (Faculty of Pharmacy, Campus Universitario de Cartuja), Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, Granada, Spain.
| | - Paul D Cotter
- VistaMilk SFI Research Centre, Cork, Ireland; Teagasc Food Research Centre, Carlow, Ireland; APC Microbiome Ireland, Cork, Ireland
| | - José A García-Salcedo
- GENYO, Centre for Genomics and Oncological Research: Pfizer / University of Granada / Andalusian Regional Government, Granada, Spain; Instituto de Investigación Biosanitaria ibs, Granada, Spain; Microbiology Unit, University Hospital Virgen de las Nieves, Granada, Spain
| |
Collapse
|
5
|
Sperling D, Rodríguez M, Guerra N, Karembe H, Diesing AK, Manso A, de Frutos L, Morales J. Simultaneous Use of Iron/Anticoccidial Treatment and Vaccination against Oedema Disease: Impact on the Development of Serum-Neutralising Antibodies, Hematinic and Anticoccidial Activities in Piglets. Vaccines (Basel) 2024; 12:1004. [PMID: 39340034 PMCID: PMC11435768 DOI: 10.3390/vaccines12091004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Oedema disease (OD) in weaned piglets is caused by shigatoxigenic Escherichia coli (STEC), which produces the Stx2e toxin. The disease is controlled by early vaccination (for example, with Ecoporc Shiga®). Iron-deficiency anaemia (IDA) and cystoisosporosis are the most common clinical conditions in piglets. These conditions are managed mainly by the intramuscular injection of iron and application of toltrazuril (for example, Forceris®). In the present study, we sought to evaluate any effect on the efficacy of OD vaccination and iron/anticoccidial treatment resulting from a simultaneous application. An evaluation was carried out by measuring the development of neutralising antibodies against the Stx2e toxin, hematinic indices and oocysts shedding. Six litters from Stx2e-antibody-negative sows were included in the study, with 12 piglets in each litter. The piglets were randomly allocated into two groups on their second day of life (DOL): (T1) iron/anticoccidial treatment and vaccine were administered on different days, and (T2) products were administered simultaneously. Blood samples were collected to determine the levels of serum-neutralising antibodies, haemoglobin and haematocrit. Faecal matter was examined for the presence of oocysts of Cystoisospora suis. No differences were found between the two groups in terms of the development of neutralising antibodies. The levels of haemoglobin and haematocrit were lower (p < 0.05 and p = 0.08, respectively) when iron/anticoccidial treatment and vaccine were applied simultaneously but within the optimal range, based on current interpretive criteria for IDA. Oocysts were not detected in the faecal samples from the animals in either group. In conclusion, we found that, under the conditions of our study, the efficacy of OD vaccination and iron/anticoccidial treatment was not affected by the simultaneous use.
Collapse
Affiliation(s)
- Daniel Sperling
- Ceva Santé Animale, 10 Avenue de la Ballastière, 33500 Libourne, France
| | - María Rodríguez
- Animal Data Analytics S.L., C/Dámaso Alonso 14, 40006 Segovia, Spain
| | - Nicolás Guerra
- Ceva Santé Animale, 10 Avenue de la Ballastière, 33500 Libourne, France
| | - Hamadi Karembe
- Ceva Santé Animale, 10 Avenue de la Ballastière, 33500 Libourne, France
| | | | - Alberto Manso
- Animal Data Analytics S.L., C/Dámaso Alonso 14, 40006 Segovia, Spain
| | - Laura de Frutos
- Animal Data Analytics S.L., C/Dámaso Alonso 14, 40006 Segovia, Spain
| | - Joaquín Morales
- Animal Data Analytics S.L., C/Dámaso Alonso 14, 40006 Segovia, Spain
| |
Collapse
|
6
|
Gao L, Xing X, Guo R, Li Q, Xu Y, Pan H, Ji P, Wang P, Yu C, Li J, An Q. Effect of Different Dietary Iron Contents on Liver Transcriptome Characteristics in Wujin Pigs. Animals (Basel) 2024; 14:2399. [PMID: 39199933 PMCID: PMC11350824 DOI: 10.3390/ani14162399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/27/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Iron is an important trace element that affects the growth and development of animals and regulates oxygen transport, hematopoiesis, and hypoxia adaptations. Wujin pig has unique hypoxic adaptability and iron homeostasis; however, the specific regulatory mechanisms have rarely been reported. This study randomly divided 18 healthy Wujin piglets into three groups: the control group, supplemented with 100 mg/kg iron (as iron glycinate); the low-iron group, no iron supplementation; and the high-iron group, supplemented with 200 mg/kg iron (as iron glycinate). The pre-feeding period was 5 days, and the formal period was 30 days. Serum was collected from empty stomachs before slaughter and at slaughter to detect changes in the serum iron metabolism parameters. Gene expression in the liver was analyzed via transcriptome analysis to determine the effects of low- and high-iron diets on transcriptome levels. Correlation analysis was performed for apparent serum parameters, and transcriptome sequencing was performed using weighted gene co-expression network analysis to reveal the key pathways underlying hypoxia regulation and iron metabolism. The main results are as follows. (1) Except for the hypoxia-inducible factor 1 (HIF-1) content (between the low- and high-iron groups), significant differences were not observed among the serum iron metabolic parameters. The serum HIF-1 content of the low-iron group was significantly higher than that of the high-iron group (p < 0.05). (2) Sequencing analysis of the liver transcriptome revealed 155 differentially expressed genes (DEGs) between the low-iron and control groups, 229 DEGs between the high-iron and control groups, and 279 DEGs between the low- and high-iron groups. Bioinformatics analysis showed that the HIF-1 and transforming growth factor-beta (TGF-β) signaling pathways were the key pathways for hypoxia regulation and iron metabolism. Four genes were selected for qPCR validation, and the results were consistent with the transcriptome sequencing data. In summary, the serum iron metabolism parameter results showed that under the influence of low- and high-iron diets, Wujin piglets maintain a steady state of physiological and biochemical indices via complex metabolic regulation of the body, which reflects their stress resistance and adaptability. The transcriptome results revealed the effects of low-iron and high-iron diets on the gene expression level in the liver and showed that the HIF-1 and TGF-β signaling pathways were key for regulating hypoxia adaptability and iron metabolism homeostasis under low-iron and high-iron diets. Moreover, HIF-1α and HEPC were the key genes. The findings provide a theoretical foundation for exploring the regulatory pathways and characteristics of iron metabolism in Wujin pigs.
Collapse
Affiliation(s)
- Lin Gao
- Yunnan Provincial Key Laboratory of Tropical and Subtropical Animal Viral Diseases, Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming 650201, China;
| | - Xiaokun Xing
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| | - Rongfu Guo
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| | - Qihua Li
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| | - Yan Xu
- Yunnan East Hunter Agriculture and Forestry Development Co., Ltd., Shuifu 657803, China;
| | - Hongbin Pan
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| | - Peng Ji
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| | - Ping Wang
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| | - Chuntang Yu
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| | - Jintao Li
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| | - Qingcong An
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| |
Collapse
|
7
|
He K, Long X, Jiang H, Qin C. The differential impact of iron on ferroptosis, oxidative stress, and inflammatory reaction in head-kidney macrophages of yellow catfish (Pelteobagrus fulvidraco) with and without ammonia stress. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 157:105184. [PMID: 38643939 DOI: 10.1016/j.dci.2024.105184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 04/23/2024]
Abstract
Ammonia toxicity in fish is closely related to ferroptosis, oxidative stress, and inflammatory responses. Iron is an essential trace element that plays a key role in many biological processes for cells and organisms, including ferroptosis, oxidative stress response, and inflammation. This study aimed to investigate the effect of iron on indicators of fish exposed to ammonia, specifically on the three aspects mentioned above. The head kidney macrophages of yellow catfish were randomly assigned to one of four groups: CON (normal control), AM (0.046 mg L-1 total ammonia nitrogen), Fe (20 μg mL-1 FeSO4), and Fe + AM (20 μg mL-1 FeSO4, 0.046 mg L-1 total ammonia nitrogen). The cells were pretreated with FeSO4 for 6 h followed by ammonia for 24 h. The study found that iron supplementation led to an excessive accumulation of iron and ROS in macrophages, but it did not strongly induce ferroptosis, oxidative stress, or inflammatory responses. This was supported by a decrease in T-AOC, and the downregulation of SOD, as well as an increase in GSH levels and the upregulation of TFR1, CAT and Nrf2. Furthermore, the mRNA expression of HIF-1, p53 and the anti-inflammatory M2 macrophage marker Arg-1 were upregulated. The results also showed that iron supplementation increased the progression of some macrophages from early apoptosis to late apoptotic cells. However, the combined treatment of iron and ammonia resulted in a stronger intracellular ferroptosis, oxidative stress, and inflammatory reaction compared to either treatment alone. Additionally, there was a noticeable increase in necrotic cells in the Fe + AM and AM groups. These findings indicate that the biological functions of iron in macrophages of fish may vary inconsistently in the presence or absence of ammonia stress.
Collapse
Affiliation(s)
- Kewei He
- Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education (Guizhou University), Guiyang, 550025, China; College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Xinran Long
- Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education (Guizhou University), Guiyang, 550025, China; College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Haibo Jiang
- Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education (Guizhou University), Guiyang, 550025, China; College of Animal Science, Guizhou University, Guiyang, 550025, China; College of Biosystems Engineering and Food Science (BEFS), Zhejiang University, Hangzhou, 310058, China.
| | - Chuanjie Qin
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, 641112, China
| |
Collapse
|
8
|
Chen X, Liu H, Liu S, Zhang Z, Li X, Mao J. Excessive dietary iron exposure increases the susceptibility of largemouth bass (Micropterus salmoides) to Aeromonas hydrophila by interfering with immune response, oxidative stress, and intestinal homeostasis. FISH & SHELLFISH IMMUNOLOGY 2024; 147:109430. [PMID: 38325595 DOI: 10.1016/j.fsi.2024.109430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/31/2024] [Accepted: 02/04/2024] [Indexed: 02/09/2024]
Abstract
Iron is an essential cofactor in the fundamental metabolic pathways of organisms. Moderate iron intake can enhance animal growth performance, while iron overload increases the risk of pathogen infection. Although the impact of iron on the pathogen-host relationship has been confirmed in higher vertebrates, research in fish is extremely limited. The effects and mechanisms of different levels of iron exposure on the infection of Aeromonas hydrophila in largemouth bass (Micropterus salmoides) remain unclear. In this study, experimental diets were prepared by adding 0, 800, 1600, and 3200 mg/kg of FeSO4∙7H2O to the basal feed, and the impact of a 56-day feeding period on the mortality rate of largemouth bass infected with A. hydrophila was analyzed. Additionally, the relationships between mortality rate and tissue iron content, immune regulation, oxidative stress, iron homeostasis, gut microbiota, and tissue morphology were investigated. The results showed that the survival rate of largemouth bass infected with A. hydrophila decreased with increasing iron exposure levels. Excessive dietary iron intake significantly increased iron deposition in the tissues of largemouth bass, reduced the expression and activity of antioxidant enzymes superoxide dismutase, catalase, and glutathione peroxidase, increased the content of lipid peroxidation product malondialdehyde, and thereby induced oxidative stress. Excessive iron supplementation could influence the immune response of largemouth bass by upregulating the expression of pro-inflammatory cytokines in the intestine and liver, while downregulating the expression of anti-inflammatory cytokines. Additionally, excessive iron intake could also affect iron metabolism by inducing the expression of hepcidin, disrupt intestinal homeostasis by interfering with the composition and function of the gut microbiota, and induce damage in the intestinal and hepatic tissues. These research findings provide a partial theoretical basis for deciphering the molecular mechanisms underlying the influence of excessive iron exposure on the susceptibility of largemouth bass to pathogenic bacteria.
Collapse
Affiliation(s)
- Xiaoli Chen
- Guangdong Engineering Research Center of High-Value Utilization and Equipment Development of Marine Biological Resources, Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, Guangdong, 511458, China
| | - Hong Liu
- College of Chemistry and Molecular Sciences, Henan University, Kaifeng, 475001, China
| | - Shuangping Liu
- Guangdong Engineering Research Center of High-Value Utilization and Equipment Development of Marine Biological Resources, Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, Guangdong, 511458, China; National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| | - Zhifeng Zhang
- Guangdong Engineering Research Center of High-Value Utilization and Equipment Development of Marine Biological Resources, Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, Guangdong, 511458, China
| | - Xiong Li
- Guangdong Engineering Research Center of High-Value Utilization and Equipment Development of Marine Biological Resources, Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, Guangdong, 511458, China
| | - Jian Mao
- Guangdong Engineering Research Center of High-Value Utilization and Equipment Development of Marine Biological Resources, Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, Guangdong, 511458, China; National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| |
Collapse
|
9
|
Zhao D, Gao Y, Chen Y, Zhang Y, Deng Y, Niu S, Dai H. L-Citrulline Ameliorates Iron Metabolism and Mitochondrial Quality Control via Activating AMPK Pathway in Intestine and Improves Microbiota in Mice with Iron Overload. Mol Nutr Food Res 2024; 68:e2300723. [PMID: 38425278 DOI: 10.1002/mnfr.202300723] [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] [Scholar Register] [Received: 10/24/2023] [Revised: 01/26/2024] [Indexed: 03/02/2024]
Abstract
SCOPE Oxidative stress caused by iron overload tends to result in intestinal mucosal barrier dysfunction and intestinal microbiota imbalance. As a neutral and nonprotein amino acid, L-Citrulline (L-cit) has been implicated in antioxidant and mitochondrial amelioration properties. This study investigates whether L-cit can alleviate iron overload-induced intestinal injury and explores the underlying mechanisms. METHODS AND RESULTS C57BL/6J mice are intraperitoneally injected with iron dextran, then gavaged with different dose of L-cit for 2 weeks. L-cit treatment significantly alleviates intestine pathological injury, oxidative stress, ATP level, and mitochondrial respiratory chain complex activities, accompanied by ameliorating mitochondrial quality control. L-cit-mediated protection is associated with the upregulation of Glutathione Peroxidase 4 (GPX4) expression, inhibition Nuclear Receptor Coactivator 4 (NCOA4)-mediated ferritinophagy and ferroptosis, and improvement of gut microbiota. To investigate the underlying molecular mechanisms, Intestinal Porcine Epithelial Cell line-J2 (IPEC-J2) cells are treated with L-cit or AMP-activated Protein Kinase (AMPK) inhibitor. AMPK signaling has been activated by L-cit. Notably, Compound C abolishes L-cit's protection on intestinal barrier, mitochondrial function, and antioxidative capacity in IPEC-J2 cells. CONCLUSION L-cit may restrain ferritinophagy and ferroptosis to regulate iron metabolism, and induce AMPK pathway activation, which contributes to exert antioxidation, ameliorate iron metabolism and mitochondrial quality control, and improve intestinal microbiota. L-cit is a promising therapeutic strategy for iron overload-induced intestinal injury.
Collapse
Affiliation(s)
- Dai Zhao
- College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Street, Hongshan District, Wuhan, Hubei, 430070, China
| | - Yuan Gao
- College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Street, Hongshan District, Wuhan, Hubei, 430070, China
| | - Yiqin Chen
- College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Street, Hongshan District, Wuhan, Hubei, 430070, China
| | - Yingsi Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Street, Hongshan District, Wuhan, Hubei, 430070, China
| | - Yian Deng
- College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Street, Hongshan District, Wuhan, Hubei, 430070, China
| | - Sai Niu
- College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Street, Hongshan District, Wuhan, Hubei, 430070, China
| | - Hanchuan Dai
- College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Street, Hongshan District, Wuhan, Hubei, 430070, China
| |
Collapse
|
10
|
Eussen SRBM, Wielders S, de Rooij WE, Van Ampting MTJ, Van Esch BCAM, de Vries JHM, Bredenoord AJ, Vlieg‐Boerstra B. Dietary composition of adult eosinophilic esophagitis patients is related to disease severity. Immun Inflamm Dis 2024; 12:e1206. [PMID: 38456617 PMCID: PMC10921897 DOI: 10.1002/iid3.1206] [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] [Scholar Register] [Received: 11/17/2023] [Revised: 02/07/2024] [Accepted: 02/15/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND In addition to the elimination diet, dietary composition may influence disease severity in patients with eosinophilic esophagitis (EoE) through modulation of the immune response. AIM To explore the immunomodulatory role of nutrition before and during elimination diet in adult EoE patients. METHODS Nutritional intake was assessed in 39 Dutch adult EoE patients participating in the Supplemental Elemental Trial (Dutch trial registry NL6014, NTR6778) using 3-day food diaries. In this randomized controlled trial, diagnosed patients received either a four-food elimination diet alone (FFED) or FFED with addition of an amino acid-based formula for 6 weeks. Multiple linear regression analyses were performed to assess associations between the intake of nutrients and food groups per 1000 kCal and peak eosinophil count/high power field (PEC), both at baseline and after 6 weeks. RESULTS At baseline, we found a statistically significant negative (thus favorable) relationship between the intake of protein, total fat, phosphorus, zinc, vitamin B12, folate, and milk products and PEC (p < .05), while calcium (p = .058) and full-fat cheese/curd (p = .056) were borderline (favorably) significant. In contrast, total carbohydrates, prepacked fruit juice, and white bread were significantly positively (unfavorable) related to PEC (p < .05), while ultra-processed meals (p = .059) were borderline (unfavorably) significant. After dietary intervention, coffee/tea were significantly negatively (favorably) related to PEC, hummus/legumes were significantly positively (unfavorably) related with PEC, while peanuts were borderline significantly positively related (p = .058). CONCLUSION Dietary composition may be related to inflammation in adult EoE patients. High-quality and anti-inflammatory diets may be a promising adjuvant therapy in the dietary management of EoE.
Collapse
Affiliation(s)
| | - Sanne Wielders
- Division of Human NutritionWageningen UniversityWageningenThe Netherlands
| | - Willemijn E. de Rooij
- Department of Gastroenterology & HepatologyAmsterdam University Medical CenterAmsterdamThe Netherlands
| | | | - Betty C. A. M. Van Esch
- Danone Nutricia ResearchUtrechtThe Netherlands
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of ScienceUtrecht UniversityUtrechtThe Netherlands
| | | | - Albert J. Bredenoord
- Department of Gastroenterology & HepatologyAmsterdam University Medical CenterAmsterdamThe Netherlands
| | | |
Collapse
|
11
|
Karamantziani T, Pouliakis A, Xanthos T, Ekmektzoglou K, Paliatsiou S, Sokou R, Iacovidou N. The Effect of Oral Iron Supplementation/Fortification on the Gut Microbiota in Infancy: A Systematic Review and Meta-Analysis. CHILDREN (BASEL, SWITZERLAND) 2024; 11:231. [PMID: 38397343 PMCID: PMC10887499 DOI: 10.3390/children11020231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024]
Abstract
(1) Background: Iron is an essential metal for the proper growth and neurodevelopment of infants. To prevent and treat iron deficiency, iron supplementation or fortification is often required. It has been shown, though, that it affects the synthesis of gut microbiota. (2) Methods: This paper is a systematic review and meta-analysis of the effect of oral iron supplementation/fortification on the gut microbiota in infancy. Studies in healthy neonates and infants who received per os iron with existing data on gut microbiota were included. Three databases were searched: PUBMED, Scopus, and Google Scholar. Randomized controlled trials (RCTs) were included. Quality appraisal was assessed using the ROB2Tool. (3) Results: A total of six RCTs met inclusion criteria for a systematic review, and four of them were included in the meta-analysis using both the fixed and random effects methods. Our results showed that there is very good heterogeneity in the iron group (I2 = 62%), and excellent heterogeneity in the non-iron group (I2 = 98%). According to the meta-analysis outcomes, there is a 10.3% (95% CI: -15.0--5.55%) reduction in the bifidobacteria population in the iron group and a -2.96% reduction for the non-iron group. There is a confirmed difference (p = 0.02) in the aggregated outcomes between iron and non-iron supplement, indicative that the bifidobacteria population is reduced when iron supplementation is given (total reduction 6.37%, 95%CI: 10.16-25.8%). (4) Conclusions: The abundance of bifidobacteria decreases when iron supplementation or fortification is given to infants.
Collapse
Affiliation(s)
- Theoni Karamantziani
- B’ Neonatal Intensive Care Unit and Neonatal High Dependency Unit, “Aghia Sofia” General Children’s Hospital, 11527 Athens, Greece;
| | - Abraham Pouliakis
- 2nd Department of Pathology, “Attikon” University Hospital, National and Kapodistrian University of Athens, 12464 Athens, Greece
| | - Theodoros Xanthos
- School of Health Sciences, University of West Attica, 12243 Athens, Greece;
| | | | - Styliani Paliatsiou
- 2nd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Aretaieio Hospital, 11528 Athens, Greece;
| | - Rozeta Sokou
- Neonatal Intensive Care Unit, “Agios Panteleimon” General Hospital of Nikea, 3 D. Mantouvalou Str., Nikea, 18454 Piraeus, Greece;
| | - Nicoletta Iacovidou
- Neonatal Department, Aretaieio Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| |
Collapse
|
12
|
Bao H, Wang Y, Xiong H, Xia Y, Cui Z, Liu L. Mechanism of Iron Ion Homeostasis in Intestinal Immunity and Gut Microbiota Remodeling. Int J Mol Sci 2024; 25:727. [PMID: 38255801 PMCID: PMC10815743 DOI: 10.3390/ijms25020727] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/01/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Iron is a vital trace element that plays an important role in humans and other organisms. It plays an active role in the growth, development, and reproduction of bacteria, such as Bifidobacteria. Iron deficiency or excess can negatively affect bacterial hosts. Studies have reported a major role of iron in the human intestine, which is necessary for maintaining body homeostasis and intestinal barrier function. Organisms can maintain their normal activities and regulate some cancer cells in the body by regulating iron excretion and iron-dependent ferroptosis. In addition, iron can modify the interaction between hosts and microorganisms by altering their growth and virulence or by affecting the immune system of the host. Lactic acid bacteria such as Lactobacillus acidophilus (L. acidophilus), Lactobacillus rhamnosus (L. rhamnosus), and Lactobacillus casei (L. casei) were reported to increase trace elements, protect the host intestinal barrier, mitigate intestinal inflammation, and regulate immune function. This review article focuses on the two aspects of the iron and gut and generally summarizes the mechanistic role of iron ions in intestinal immunity and the remodeling of gut microbiota.
Collapse
Affiliation(s)
| | | | | | | | - Zhifu Cui
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; (H.B.); (Y.W.); (H.X.); (Y.X.)
| | - Lingbin Liu
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; (H.B.); (Y.W.); (H.X.); (Y.X.)
| |
Collapse
|
13
|
Liu Y, Li G, Lu F, Guo Z, Cai S, Huo T. Excess iron intake induced liver injury: The role of gut-liver axis and therapeutic potential. Biomed Pharmacother 2023; 168:115728. [PMID: 37864900 DOI: 10.1016/j.biopha.2023.115728] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/23/2023] Open
Abstract
Excessive iron intake is detrimental to human health, especially to the liver, which is the main organ for iron storage. Excessive iron intake can lead to liver injury. The gut-liver axis (GLA) refers to the bidirectional relationship between the gut and its microbiota and the liver, which is a combination of signals generated by dietary, genetic and environmental factors. Excessive iron intake disrupts the GLA at multiple interconnected levels, including the gut microbiota, gut barrier function, and the liver's innate immune system. Excessive iron intake induces gut microbiota dysbiosis, destroys gut barriers, promotes liver exposure to gut microbiota and its derived metabolites, and increases the pro-inflammatory environment of the liver. There is increasing evidence that excess iron intake alters the levels of gut microbiota-derived metabolites such as secondary bile acids (BAs), short-chain fatty acids, indoles, and trimethylamine N-oxide, which play an important role in maintaining homeostasis of the GLA. In addition to iron chelators, antioxidants, and anti-inflammatory agents currently used in iron overload therapy, gut barrier intervention may be a potential target for iron overload therapy. In this paper, we review the relationship between excess iron intake and chronic liver diseases, the regulation of iron homeostasis by the GLA, and focus on the effects of excess iron intake on the GLA. It has been suggested that probiotics, fecal microbiota transfer, farnesoid X receptor agonists, and microRNA may be potential therapeutic targets for iron overload-induced liver injury by protecting gut barrier function.
Collapse
Affiliation(s)
- Yu Liu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China
| | - Guangyan Li
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China
| | - Fayu Lu
- School of Public Health, China Medical University, Shenyang, Liaoning 110122, China
| | - Ziwei Guo
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China
| | - Shuang Cai
- The First Affiliated Hospital of China Medical University, Shenyang 110001, China.
| | - Taoguang Huo
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
14
|
Dong Z, Liu S, Deng Q, Li G, Tang Y, Wu X, Wan D, Yin Y. Role of iron in host-microbiota interaction and its effects on intestinal mucosal growth and immune plasticity in a piglet model. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2086-2098. [PMID: 37530911 DOI: 10.1007/s11427-022-2409-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/15/2023] [Indexed: 08/03/2023]
Abstract
Iron is an essential trace element for both the host and resident microbes in the gut. In this study, iron was administered orally and parenterally to anemic piglets to investigate the role of iron in host-microbiota interaction and its effects on intestinal mucosal growth and immune plasticity. We found that oral iron administration easily increased the abundance of Proteobacteria and Escherichia-Shigella, and decreased the abundance of Lactobacillus in the ileum. Furthermore, similar bacterial changes, namely an increase in Proteobacteria, Escherichia-Shigella, and Fusobacterium and a reduction in the Christensenellaceae_R-7_group, were observed in the colon of both iron-supplemented groups. Spearman's correlation analysis indicated that the changed Fusobacterium, Fusobacteria and Proteobacteria in the colon were positively correlated with hemoglobin, colon and spleen iron levels. Nevertheless, it was found that activated mTOR1 signaling, improved villous height and crypt depth in the ileum, enhanced immune communication, and increased protein expression of IL-22 and IL-10 in the colon of both iron-supplemented groups. In conclusion, the benefits of improved host iron outweigh the risks of altered gut microbiota for intestinal mucosal growth and immune regulation in treating iron deficiency anemia.
Collapse
Affiliation(s)
- Zhenglin Dong
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Shuan Liu
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Qingqing Deng
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Guanya Li
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Yulong Tang
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Xin Wu
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Dan Wan
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| | - Yulong Yin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| |
Collapse
|
15
|
Liu S, Dong Z, Tang W, Zhou J, Guo L, Gong C, Liu G, Wan D, Yin Y. Dietary iron regulates intestinal goblet cell function and alleviates Salmonella typhimurium invasion in mice. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2006-2019. [PMID: 37340176 DOI: 10.1007/s11427-022-2298-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 01/31/2023] [Indexed: 06/22/2023]
Abstract
Iron is an important micronutrient that plays a vital role in host defenses and bacterial pathogenicity. As iron treatments increase the risk of infection by stimulating the growth and virulence of bacterial pathogens, their roles in anti-infection immunity have frequently been underestimated. To estimate whether adequate dietary iron intake would help defend against pathogenic bacterial infection, mice were fed iron-deficient (2 mg kg-1 feed), iron-sufficient (35 mg kg-1 feed), or iron-enriched diet (350 mg kg-1 feed) for 12 weeks, followed by oral infection with Salmonella typhimurium. Our results revealed that dietary iron intake improved mucus layer function and decelerated the invasion of the pathogenic bacteria, Salmonella typhimurium. Positive correlations between serum iron and the number of goblet cells and mucin2 were found in response to total iron intake in mice. Unabsorbed iron in the intestinal tract affected the gut microbiota composition, and the abundance of Bacteroidales, family Muribaculaceae, was positively correlated with their mucin2 expression. However, the results from antibiotic-treated mice showed that the dietary iron-regulated mucin layer function was not microbial-dependent. Furthermore, in vitro studies revealed that ferric citrate directly induced mucin2 expression and promoted the proliferation of goblet cells in both ileal and colonic organoids. Thus, dietary iron intake improves serum iron levels, regulates goblet cell regeneration and mucin layer function, and plays a positive role in the prevention of pathogenic bacteria.
Collapse
Affiliation(s)
- Shuan Liu
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Zhenlin Dong
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Wenjie Tang
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, 610066, China
| | - Jian Zhou
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Liu Guo
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Chengyan Gong
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Guang Liu
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Dan Wan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- University of Chinese Academy of Sciences, Beijing, 101408, China.
| |
Collapse
|
16
|
Zeng Y, Zhou B, Huang L, Liu Y. Iron-rich Candida utilis improves intestinal health in weanling piglets. J Appl Microbiol 2023; 134:lxad135. [PMID: 37401152 DOI: 10.1093/jambio/lxad135] [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] [Scholar Register] [Received: 03/29/2023] [Revised: 06/17/2023] [Accepted: 06/30/2023] [Indexed: 07/05/2023]
Abstract
AIM This study aimed to investigate the effects of substituting inorganic iron in the diet of weanling piglets with iron-rich Candida utilis on gut morphology, immunity, barrier, and microbiota. METHODS AND RESULTS Seventy-two healthy 28-day-old Duroc × Landrace × Yorkshire desexed male weanling piglets were randomly assigned to 2 groups (n = 6), with 6 pens per group and 6 piglets in each pen. The control group was fed a basal diet containing ferrous sulfate (104 mg kg-1 iron), while the experimental group was fed a basal diet supplemented with iron-rich C. utilis (104 mg kg-1 iron). The results show that the growth performance of weanling piglets showed no significantly differences (P > 0.05). Iron-rich C. utilis significantly elevated villus height and decreased crypt depth in the duodenum and jejunum (P < 0.05). Additionally, there was a significant increase in SIgA content, a down-regulated of pro-inflammatory factors expression, and an up-regulated of anti-inflammatory factors expression in the jejunum and ileum of piglets fed iron-rich C. utilis (P < 0.05). The mRNA expression levels of ZO-1, Claudin-1, Occludin, and Mucin2 in the jejunum were significantly increased by iron-rich C. utilis, and were significantly increased ZO-1 and Claudin-1 in the ileum (P < 0.05). The colonic microbiota, however, was not significantly affected by iron-rich C. utilis (P > 0.05). CONCLUSION Iron-rich C. utilis improved intestinal morphology and structure, as well as intestinal immunity and intestinal barrier function.
Collapse
Affiliation(s)
- Yan Zeng
- Hunan Institute of Microbiology, Changsha 410009, China
| | - Bingyu Zhou
- Hunan Institute of Microbiology, Changsha 410009, China
| | - LiHong Huang
- Hunan Institute of Microbiology, Changsha 410009, China
| | - YuBo Liu
- Hunan Institute of Microbiology, Changsha 410009, China
| |
Collapse
|
17
|
Sciascia QL, Metges CC. Review: Methods and biomarkers to investigate intestinal function and health in pigs. Animal 2023; 17 Suppl 3:100860. [PMID: 37316380 DOI: 10.1016/j.animal.2023.100860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 06/16/2023] Open
Abstract
Society is becoming increasingly critical of animal husbandry due to its environmental impact and issues involving animal health and welfare including scientific experiments conducted on farm animals. This opens up two new fields of scientific research, the development of non- or minimally invasive (1) methods and techniques using faeces, urine, breath or saliva sampling to replace existing invasive models, and (2) biomarkers reflecting a disease or malfunction of an organ that may predict the future outcome of a pig's health, performance or sustainability. To date, there is a paucity of non- or minimally invasive methods and biomarkers investigating gastrointestinal function and health in pigs. This review describes recent literature pertaining to parameters that assess gastrointestinal functionality and health, tools currently used to investigate them, and the development or the potential to develop new non- and minimally invasive methods and/or biomarkers in pigs. Methods described within this review are those that characterise gastrointestinal mass such as the citrulline generation test, intestinal protein synthesis rate, first pass splanchnic nutrient uptake and techniques describing intestinal proliferation, barrier function and transit rate, and microbial composition and metabolism. An important consideration is gut health, and several molecules with the potential to act as biomarkers of compromised gut health in pigs are reported. Many of these methods to investigate gut functionality and health are considered 'gold standards' but are invasive. Thus, in pigs, there is a need to develop and validate non-invasive methods and biomarkers that meet the principles of the 3 R guidelines, which aim to reduce and refine animal experimentation and replace animals where possible.
Collapse
Affiliation(s)
- Q L Sciascia
- Research Institute for Farm Animal Biology, Institute of Nutritional Physiology "Oskar Kellner", Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - C C Metges
- Research Institute for Farm Animal Biology, Institute of Nutritional Physiology "Oskar Kellner", Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany.
| |
Collapse
|
18
|
Wang L, Wang C, Peng Y, Zhang Y, Liu Y, Liu Y, Yin Y. Research progress on anti-stress nutrition strategies in swine. ANIMAL NUTRITION 2023; 13:342-360. [DOI: 10.1016/j.aninu.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/04/2023] [Accepted: 03/30/2023] [Indexed: 04/09/2023]
|
19
|
Thilakarathna WPDW, Langille MGI, Rupasinghe HPV. Hepatotoxicity of polymeric proanthocyanidins is caused by translocation of bacterial lipopolysaccharides through impaired gut epithelium. Toxicol Lett 2023; 379:35-47. [PMID: 36935082 DOI: 10.1016/j.toxlet.2023.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/26/2023] [Accepted: 03/08/2023] [Indexed: 03/19/2023]
Abstract
Polymeric proanthocyanidins (P-PAC) induced hepatotoxicity in C57BL/6 mice. Mice were supplemented with P-PAC alone or with a mixture of probiotic bacteria (PB), Lactobacillus, Bifidobacterium, and Akkermansia muciniphila for 14 consecutive days. The liver tissues of sacrificed mice were analyzed by mass spectrometry to identify and quantify the P-PAC metabolites. Potential P-PAC metabolites, 2-hydroxyphenylacetic acid and pyrocatechol were detected in higher concentrations and 4-hydroxybenzoic acid was detected exclusively in the mice supplemented with P-PAC and PB. Supplementation with P-PAC alone or with PB caused no shift in the α-diversity of mice gut microbiota. P-PAC induced nonalcoholic steatohepatitis in mice through increasing liver exposure to intestinal bacterial lipopolysaccharides by reducing expression of gut epithelial tight junction proteins, claudin-3 and occludin. Lipopolysaccharide concentrations in the livers of mice supplemented with P-PAC were significantly high compared to the control mice. Furthermore, P-PAC downregulated the expressions of claudin-3 and claudin-4 tight junction proteins in cultured Caco-2 cell monolayers. PB biotransformed P-PAC into bioavailable metabolites and potentially reduced the toxicity of P-PAC. The toxicity of P-PAC and their synbiotics need to be critically evaluated for the safety of human consumption.
Collapse
Affiliation(s)
- Wasitha P D W Thilakarathna
- Department of Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, Nova Scotia, Canada
| | - Morgan G I Langille
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - H P Vasantha Rupasinghe
- Department of Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, Nova Scotia, Canada; Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
20
|
Ortín-Bustillo A, Escribano D, Martínez-Subiela S, Tvarijonaviciute A, Muñoz-Prieto A, López-Arjona M, Cerón JJ, Tecles F. Trace Elements and Ferritin in Pig Saliva: Variations during Fattening, Time of Sampling, Effect of Dirtiness and Stability under Different Storage Conditions. Antioxidants (Basel) 2023; 12:antiox12030649. [PMID: 36978897 PMCID: PMC10045741 DOI: 10.3390/antiox12030649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
The objective of this study was to evaluate the possible changes of zinc (Zn), copper (Cu), iron (Fe) and ferritin during the entire productive cycle in fattening pigs and at different diurnal sampling times. Moreover, the possible effects of the presence of pen contaminants and storage stability at different temperature conditions were assessed. The analytes changed along the different phases of the fattening productive cycle, showing, in general, higher values at the initial phases. In addition, statistically significant variations were found in Zn and Cu measurements at different sampling times of the day. In the spectrophotometric assays, the values of all analytes significantly increased after adding high concentrations of feces or feed. However, when low concentrations of feces or feed were added, only Cu showed a significant increase. Overall, the salivary levels of Zn, Cu, Fe and ferritin in pigs can change during different fattening phases and the different hours of the day. These analytes were more stable at −80 °C and, if saliva is contaminated with feces or feed, it can lead to an increase in these analytes.
Collapse
Affiliation(s)
- Alba Ortín-Bustillo
- Interdisciplinary Laboratory of Clinical Analysis (Interlab-UMU), Regional Campus of International Excellence ‘Campus Mare Nostrum’, University of Murcia, Campus de Espinardo s/n, 30100 Murcia, Spain
| | - Damián Escribano
- Interdisciplinary Laboratory of Clinical Analysis (Interlab-UMU), Regional Campus of International Excellence ‘Campus Mare Nostrum’, University of Murcia, Campus de Espinardo s/n, 30100 Murcia, Spain
- Department of Animal Production, Regional Campus of International Excellence ‘Campus Mare Nostrum’, University of Murcia, Campus de Espinardo s/n, 30100 Murcia, Spain
| | - Silvia Martínez-Subiela
- Interdisciplinary Laboratory of Clinical Analysis (Interlab-UMU), Regional Campus of International Excellence ‘Campus Mare Nostrum’, University of Murcia, Campus de Espinardo s/n, 30100 Murcia, Spain
| | - Asta Tvarijonaviciute
- Interdisciplinary Laboratory of Clinical Analysis (Interlab-UMU), Regional Campus of International Excellence ‘Campus Mare Nostrum’, University of Murcia, Campus de Espinardo s/n, 30100 Murcia, Spain
| | - Alberto Muñoz-Prieto
- Interdisciplinary Laboratory of Clinical Analysis (Interlab-UMU), Regional Campus of International Excellence ‘Campus Mare Nostrum’, University of Murcia, Campus de Espinardo s/n, 30100 Murcia, Spain
| | - Marina López-Arjona
- Department of Animal and Food Science, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - José J. Cerón
- Interdisciplinary Laboratory of Clinical Analysis (Interlab-UMU), Regional Campus of International Excellence ‘Campus Mare Nostrum’, University of Murcia, Campus de Espinardo s/n, 30100 Murcia, Spain
| | - Fernando Tecles
- Interdisciplinary Laboratory of Clinical Analysis (Interlab-UMU), Regional Campus of International Excellence ‘Campus Mare Nostrum’, University of Murcia, Campus de Espinardo s/n, 30100 Murcia, Spain
- Correspondence: ; Tel.: +34-868-887-082
| |
Collapse
|
21
|
The Impacts of Iron Overload and Ferroptosis on Intestinal Mucosal Homeostasis and Inflammation. Int J Mol Sci 2022; 23:ijms232214195. [PMID: 36430673 PMCID: PMC9697168 DOI: 10.3390/ijms232214195] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022] Open
Abstract
Intestinal homeostasis is maintained through the interplay of the intestinal mucosa, local and systemic immune factors, and the microbial content of the gut. Iron is a trace mineral in most organisms, including humans, which is essential for growth, systemic metabolism and immune response. Paradoxically, excessive iron intake and/or high iron status can be detrimental to iron metabolism in the intestine and lead to iron overload and ferroptosis-programmed cell death mediated by iron-dependent lipid peroxidation within cell membranes, which contributes to several intestinal diseases. In this review, we comprehensively review recent findings on the impacts of iron overload and ferroptosis on intestinal mucosal homeostasis and inflammation and then present the progress of iron overload and ferroptosis-targeting therapy in intestinal diseases. Understanding the involved mechanisms can provide a new understanding of intestinal disease pathogenesis and facilitate advanced preventive and therapeutic strategies for intestinal dysfunction and diseases.
Collapse
|
22
|
The Effects and Cell Barrier Mechanism of Main Dietary Nutrients on Intestinal Barrier. Curr Opin Food Sci 2022. [DOI: 10.1016/j.cofs.2022.100942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
23
|
Malesza IJ, Bartkowiak-Wieczorek J, Winkler-Galicki J, Nowicka A, Dzięciołowska D, Błaszczyk M, Gajniak P, Słowińska K, Niepolski L, Walkowiak J, Mądry E. The Dark Side of Iron: The Relationship between Iron, Inflammation and Gut Microbiota in Selected Diseases Associated with Iron Deficiency Anaemia—A Narrative Review. Nutrients 2022; 14:nu14173478. [PMID: 36079734 PMCID: PMC9458173 DOI: 10.3390/nu14173478] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 12/21/2022] Open
Abstract
Iron is an indispensable nutrient for life. A lack of it leads to iron deficiency anaemia (IDA), which currently affects about 1.2 billion people worldwide. The primary means of IDA treatment is oral or parenteral iron supplementation. This can be burdened with numerous side effects such as oxidative stress, systemic and local-intestinal inflammation, dysbiosis, carcinogenic processes and gastrointestinal adverse events. Therefore, this review aimed to provide insight into the physiological mechanisms of iron management and investigate the state of knowledge of the relationship between iron supplementation, inflammatory status and changes in gut microbiota milieu in diseases typically complicated with IDA and considered as having an inflammatory background such as in inflammatory bowel disease, colorectal cancer or obesity. Understanding the precise mechanisms critical to iron metabolism and the awareness of serious adverse effects associated with iron supplementation may lead to the provision of better IDA treatment. Well-planned research, specific to each patient category and disease, is needed to find measures and methods to optimise iron treatment and reduce adverse effects.
Collapse
Affiliation(s)
- Ida J. Malesza
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | | | - Jakub Winkler-Galicki
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Aleksandra Nowicka
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | | | - Marta Błaszczyk
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Paulina Gajniak
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Karolina Słowińska
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Leszek Niepolski
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Jarosław Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Edyta Mądry
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
- Correspondence:
| |
Collapse
|
24
|
Zhou N, Tian Y, Liu W, Tu B, Xu W, Gu T, Zou K, Lu L. Protective Effects of Resveratrol and Apigenin Dietary Supplementation on Serum Antioxidative Parameters and mRNAs Expression in the Small Intestines of Diquat-Challenged Pullets. Front Vet Sci 2022; 9:850769. [PMID: 35711792 PMCID: PMC9196582 DOI: 10.3389/fvets.2022.850769] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Poultry as a large-scale intensive farming is vulnerable to oxidative stress. Resveratrol and apigenin are recognized to have many beneficial bioactive functions. This study tested the hypothesis that dietary resveratrol and apigenin supplementation alleviates oxidative stress in the small intestine of diquat-challenged pullets. A total of 200 healthy pullets were randomly divided into four treatment groups: control group fed with a basal diet (CON), diquat group fed with a basal diet (DIQ), resveratrol group fed with a basal diet containing 500 mg/kg resveratrol (RES), and an apigenin group fed with a basal diet containing 500 mg/kg apigenin (API) and injected intraperitoneally with either 1 ml of saline (CON) or 8 mg/kg body weight of diquat (DIQ, RES, and API) to induce oxidative stress. The day of the injection was considered as day 0. The results indicated that resveratrol and apigenin were able to decrease the malondialdehyde (MDA) level and upregulate total antioxidant capacity (T-AOC), superoxide dismutase (SOD), and glutathione peroxidase (GSH-PX) levels in serum on day 1 and 10 after being diquat-challenged. In addition, resveratrol increased mRNA expression of NQO1 (NAD(P)H dehydrogenase quinone 1) and HO-1 (heme oxygenase-1) in ileum and jejunum on day 10, while apigenin upregulated nuclear factor erythroid 2-related factor 2 (NRF2), NQO1, and HO-1 in ileum and jejunum on day 10. Both resveratrol and apigenin increased the mRNA expression of CLAUDIN-1 in ileum and jejunum on day 1 and that of ZO-1 (zonula occludens-1) in ileum on day 10 post-diquat-injection. These findings indicate that dietary supplementation with resveratrol and apigenin attenuates oxidative stress involving NRF2 signaling pathways in diquat-challenged pullets to some extent. These observations are valuable for the chicken industry and resveratrol and apigenin applications in animal husbandry.
Collapse
Affiliation(s)
- Ning Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science and Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Yong Tian
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science and Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Wenchao Liu
- Huzhou Lvchang Ecoagriculture Co., Ltd., Huzhou, China
| | - Bingjiang Tu
- Animal Disease Prevention and Control Center, Huzhou Wuxing District Agricultural and Rural Bureau, Huzhou, China
| | - Wenwu Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science and Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Tiantian Gu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science and Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Kang Zou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Kang Zou
| | - Lizhi Lu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science and Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
- Lizhi Lu
| |
Collapse
|
25
|
Zhou N, Tian Y, Liu W, Tu B, Gu T, Xu W, Zou K, Lu L. Effects of quercetin and coated sodium butyrate dietary supplementation in diquat-challenged pullets. Anim Biosci 2022; 35:1434-1443. [PMID: 35240016 PMCID: PMC9449397 DOI: 10.5713/ab.21.0493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/02/2022] [Indexed: 11/27/2022] Open
Abstract
Objective This study was designed to investigate the hypothesis that dietary quercetin (QUE) and coated sodium butyrate (SB) supplementation alleviate oxidative stress in the small intestine of diquat (DIQ)-challenged pullets. Methods A total of 200 13-week-old pullets were divided into four groups: the control group (CON), the DIQ group, the QUE group, and the coated SB group, and injected intraperitoneally with either saline (CON) or diquat (DIQ, QUE, and SB) to induce oxidative stress on day 0. Results On the first day, the malondialdehyde and superoxide dismutase (SOD) concentrations in the SB group were significantly different from those in the DIQ and QUE groups (p<0.05), and dietary supplementation with SB increased serum glutathione peroxidase (GSH-PX) levels compared with the DIQ group (p<0.05). Quercetin and SB increased the levels of CLAUDIN-1 and zonula occludens-1 (ZO-1) in the jejunum. On the tenth day of treatment, QUE attenuated the decrease in GSH-PX levels compared to those of the CON group (p<0.05), while SB increased SOD, GSH-PX, and total antioxidant capacity levels compared to those of the DIQ group. Nuclear factor erythroid 2-related factor 2 (NRF2) and heme oxygenase-1 (HO-1) mRNA levels in the QUE and SB groups increased (p<0.05) and CLAUDIN-1 mRNA levels in the QUE and SB groups were upregulated compared to those in the DIQ group ileum tissue. Conclusion Supplementation of QUE and SB demonstrated the ability to relieve oxidative stress in pullets post DIQ-injection with a time-dependent manner and QUE and SB may be potential antioxidant additives for relieving oxidative stress and protecting the intestinal barrier of pullets.
Collapse
Affiliation(s)
- Ning Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.,State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310000, China
| | - Yong Tian
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310000, China
| | - Wenchao Liu
- Huzhou Lvchang Ecoagriculture Co., Ltd. Huzhou, 313025, China
| | - Bingjiang Tu
- Huzhou Wuxing District Animal Disease Prevention and Control Center, Huzhou, 313000, China
| | - Tiantian Gu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310000, China
| | - Wenwu Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310000, China
| | - Kang Zou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Lizhi Lu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.,State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310000, China
| |
Collapse
|
26
|
Effect of Lacticaseibacillus casei Zhang on iron status, immunity, and gut microbiota of mice fed with low-iron diet. J Funct Foods 2022. [DOI: 10.1016/j.jff.2021.104906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
27
|
Insight into the Pro-inflammatory and Profibrotic Role of Macrophage in Heart Failure With Preserved Ejection Fraction. J Cardiovasc Pharmacol 2021; 76:276-285. [PMID: 32501838 DOI: 10.1097/fjc.0000000000000858] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The prevalence of heart failure (HF) with preserved ejection fraction (HFpEF) is higher than that of HF with reduced/midrange ejection fraction (HFrEF/HFmrEF). However, no evidence-based guidelines for managing HFpEF have been generated. The current body of knowledge indicates that fibrosis and inflammation are important components of the cardiac remodeling process in HFpEF. In addition, macrophages potentially play an important role in pro-inflammatory and profibrotic processes in HFpEF patients, whereas HFpEF comorbidities could be a driving force for systemic microvascular inflammation and endothelial dysfunction. Under such circumstances, macrophages reportedly contribute to inflammation and fibrosis through 3 phases namely, inflammation, repair, and resolution. Signal transduction pathway-targeted therapies using animal experiments have generated important discoveries and breakthroughs for understanding the underlying mechanisms of HFpEF. However, only a handful of studies have reported promising results using human trials. Further investigations are therefore needed to elucidate the exact mechanisms underlying HFpEF and immune-pathogenesis of cardiac fibrosis.
Collapse
|
28
|
Dong Z, Li L, Zhang Y, Guo L, Wu X, Yin Y, Wan D. Effects of circadian iron administration on iron bioavailability and biological rhythm in pigs. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:2712-2717. [PMID: 33124038 DOI: 10.1002/jsfa.10897] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/15/2020] [Accepted: 10/30/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Iron supplements are limited by their poor absorption and low efficacy. A circadian feeding schedule would affect the circadian rhythm and improve nutrient metabolism. In this study, 18 iron-deficient piglets were randomly assigned to three groups: a control group receiving a constant diet with mid-iron (MI), a 'HL' group receiving a high-iron (HI) diet at 8:00 h and a low-iron (LI) diet at 18:00, and an 'LH' group receiving a LI diet at 8:00 and a HI diet at 18:00. The effects of circadian iron administration on iron absorption, iron status, and biological rhythm in iron-deficient piglets were investigated. RESULTS Serum iron and hemoglobin improved significantly (P < 0.05) but did not significantly differ in the circadian iron-feeding groups (P > 0.05). Iron concentration in the liver and spleen was significantly higher in the LH group than in the HL group (P < 0.05), and mRNA expression of divalent metal transport 1 (DMT1), cytochrome B (CYBRD1) and ferroportin (FPN) genes in the duodenum was significantly elevated in the LH group (P < 0.05). The clock-related genes showed differential expression in the duodenum, with greater mRNA expression for period (Per2) and cryptochrome (Cry1 and Cry2) in the LH group (P < 0.05). CONCLUSION Circadian iron administration affected iron absorption and iron storage in pigs. Iron supplementation in the evening might be a more effective pattern for iron utilization. The rhythmic system in the intestine, driven by the time, played an important role in this process. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zhenglin Dong
- Key Laboratory of Agro-Ecological Processess in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan international joint laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Lan Li
- Key Laboratory of Agro-Ecological Processess in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan international joint laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yiming Zhang
- Key Laboratory of Agro-Ecological Processess in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan international joint laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Liu Guo
- Key Laboratory of Agro-Ecological Processess in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
| | - Xin Wu
- Key Laboratory of Agro-Ecological Processess in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
| | - Yulong Yin
- Key Laboratory of Agro-Ecological Processess in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan international joint laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Dan Wan
- Key Laboratory of Agro-Ecological Processess in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
| |
Collapse
|
29
|
Luo Q, Lao C, Huang C, Xia Y, Ma W, Liu W, Chen Z. Iron Overload Resulting from the Chronic Oral Administration of Ferric Citrate Impairs Intestinal Immune and Barrier in Mice. Biol Trace Elem Res 2021; 199:1027-1036. [PMID: 32468223 DOI: 10.1007/s12011-020-02218-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/22/2020] [Indexed: 01/15/2023]
Abstract
Ferric citrate (FC) is an iron-containing phosphate binder used as a food additive for iron supplementation. To explore the potential effect of ferric citrate on intestinal epithelial function, in the present study, we administered the mice orally for 16 weeks with different doses of iron citrate (2.5 mg/day (1.25%), 5 mg/day (2.5%), and 10 mg/day (5.0%)). We found that the iron levels of serum and tissue significantly increased, which caused the body to be in an iron overload state; meanwhile, the villus height, the ratio of villus height to crypt depth, and the number of intraepithelial lymphocytes and goblet cells in jejunum all decreased. Iron overload upregulated the pro-inflammatory cytokines (IL-1β, IL-2, IL-6, TNF-ɑ), while downregulated the anti-inflammatory cytokines (IL-4, IL-10) and sIgA. Moreover, iron overload increased serum D-lactate (D-LA) levels and decreased tight junction proteins (claudin-1, occludin, and ZO-1), MUC-2, and TFF3. In addition, iron overload upregulated the content of MDA and protein carbonyl, while downregulated the activity and content of T-AOC, GSH-PX, SOD, CAT, and GSH. To sum up, the present results showed that long-term oral administration of FC resulted in iron overload, which consequently impaired intestinal immune and barrier function in mice. Meanwhile, the effect on intestinal damage may be highly related to the increase of oxidative stress in the jejunum.
Collapse
Affiliation(s)
- Qihui Luo
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Chengjie Lao
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Chao Huang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yu Xia
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Wenjing Ma
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Wentao Liu
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhengli Chen
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
30
|
Perng V, Li C, Klocke CR, Navazesh SE, Pinneles DK, Lein PJ, Ji P. Iron Deficiency and Iron Excess Differently Affect Dendritic Architecture of Pyramidal Neurons in the Hippocampus of Piglets. J Nutr 2021; 151:235-244. [PMID: 33245133 DOI: 10.1093/jn/nxaa326] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/13/2020] [Accepted: 10/01/2020] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Both iron deficiency and overload may adversely affect neurodevelopment. OBJECTIVES The study assessed how changes in early-life iron status affect iron homeostasis and cytoarchitecture of hippocampal neurons in a piglet model. METHODS On postnatal day (PD) 1, 30 Hampshire × Yorkshire crossbreed piglets (n = 15/sex) were stratified by sex and litter and randomly assigned to experimental groups receiving low (L-Fe), adequate (A-Fe), or high (H-Fe) levels of iron supplement during the pre- (PD1-21) and postweaning periods (PD22-35). Pigs in the L-Fe, A-Fe, and H-Fe groups orally received 0, 1, and 30 mg Fe · kg weight-1 · d-1 preweaning and were fed a diet containing 30, 125, and 1000 mg Fe/kg postweaning, respectively. Heme indexes were analyzed weekly, and gene and protein expressions of iron regulatory proteins in duodenal mucosa, liver, and hippocampus were analyzed through qRT-PCR and western blot, respectively, on PD35. Hippocampal neurons stained using the Golgi-Cox method were traced and their dendritic arbors reconstructed in 3-D using Neurolucida. Dendritic complexity was quantified using Sholl and branch order analyses. RESULTS Pigs in the L-Fe group developed iron deficiency anemia (hemoglobin = 8.2 g/dL, hematocrit = 20.1%) on PD35 and became stunted during week 5 with lower final body weight than H-Fe group pigs (6.6 compared with 9.6 kg, P < 0.05). In comparison with A-Fe, H-Fe increased hippocampal ferritin expression by 38% and L-Fe decreased its expression by 52% (P < 0.05), suggesting altered hippocampal iron stores. Pigs in the H-Fe group had greater dendritic complexity in CA1/3 pyramidal neurons than L-Fe group pigs as shown by more dendritic intersections with Sholl rings (P ≤ 0.04) and a greater number of dendrites (P ≤ 0.016). CONCLUSIONS In piglets, the developing hippocampus is susceptible to perturbations by dietary iron, with deficiency and overload differentially affecting dendritic arborization.
Collapse
Affiliation(s)
- Vivian Perng
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Chong Li
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Carolyn R Klocke
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA
| | - Shya E Navazesh
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Danna K Pinneles
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA
| | - Peng Ji
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| |
Collapse
|
31
|
Chen S, Wu X, Wang X, Shao Y, Tu Q, Yang H, Yin J, Yin Y. Responses of Intestinal Microbiota and Immunity to Increasing Dietary Levels of Iron Using a Piglet Model. Front Cell Dev Biol 2020; 8:603392. [PMID: 33392192 PMCID: PMC7773786 DOI: 10.3389/fcell.2020.603392] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022] Open
Abstract
Iron is an essential metal for both animals and microbiota. In general, neonates and infants of humans and animals are at the risk of iron insufficiency. However, excess dietary iron usually causes negative impacts on the host and microbiota. This study aimed to investigate overloaded dietary iron supplementation on growth performance, the distribution pattern of iron in the gut lumen and the host, intestinal microbiota, and intestine transcript profile of piglets. Sixty healthy weaning piglets were randomly assigned to six groups: fed on diets supplemented with ferrous sulfate monohydrate at the dose of 50 ppm (Fe50 group), 100 ppm (Fe100 group), 200 ppm (Fe200 group), 500 ppm (Fe500 group), and 800 ppm (Fe800), separately, for 3 weeks. The results indicated that increasing iron had no significant effects on growth performance, but increased diarrheal risk and iron deposition in intestinal digesta, tissues of intestine and liver, and serum. High iron also reduced serum iron-binding capacity, apolipoprotein, and immunoglobin A. The RNA-sequencing analysis revealed that iron changed colonic transcript profile, such as interferon gamma-signal transducer and activator of transcription two-based anti-infection gene network. Increasing iron also shifted colonic and cecal microbiota, such as reducing alpha diversity and the relative abundance of Clostridiales and Lactobacillus reuteri and increasing the relative abundance of Lactobacillus and Lactobacillus amylovorus. Collectively, this study demonstrated that high dietary iron increased diarrheal incidence, changed intestinal immune response-associated gene expression, and shifted gut microbiota. The results would enhance our knowledge of iron effects on the gut and microbiome in piglets and further contribute to understanding these aspects in humans.
Collapse
Affiliation(s)
- Shuai Chen
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Xin Wu
- College of Animal Science and Technology, Hunan Agriculture University, Hunan Co-Innovation Center of Animal Production Safety, Changsha, China
| | - Xia Wang
- College of Animal Science and Technology, Hunan Agriculture University, Hunan Co-Innovation Center of Animal Production Safety, Changsha, China.,Yiyang Vocational Technical College, Yiyang, China
| | - Yirui Shao
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Qiang Tu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Huansheng Yang
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,Animal Nutrition and Human Health Laboratory, School of Life Sciences, Hunan Normal University, Changsha, China
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agriculture University, Hunan Co-Innovation Center of Animal Production Safety, Changsha, China
| | - Yulong Yin
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Animal Science and Technology, Hunan Agriculture University, Hunan Co-Innovation Center of Animal Production Safety, Changsha, China
| |
Collapse
|
32
|
Williams HE, DeRouchey JM, Woodworth JC, Dritz SS, Tokach MD, Goodband RD, Holtcamp AJ, Bortoluzzi EM, Gebhardt JT. Effects of increasing Fe dosage in newborn pigs on suckling and subsequent nursery performance and hematological and immunological criteria. J Anim Sci 2020; 98:5871432. [PMID: 32663303 DOI: 10.1093/jas/skaa221] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/09/2020] [Indexed: 12/12/2022] Open
Abstract
A total of 336 newborn pigs (DNA 241 × 600, initially 1.75 ± 0.05 kg bodyweight [BW]) from 28 litters were used in a 63-d study evaluating the effects of increasing injectable Fe dose on suckling and subsequent nursery pig performance and blood Fe status. GleptoForte (Ceva Animal Health, LLC, Lenexa, KS) contains gleptoferron which is an Fe macromolecule complex that is commercially used as an injectable Fe source for suckling piglets. On the day of processing (day 3 after birth), all piglets were weighed and 6 barrows and 6 gilts per litter were allotted within sex to 1 of 6 treatments in a completely randomized design. Treatments consisted of a negative control receiving no Fe injection and increasing injectable Fe to achieve either 50, 100, 150, 200 mg, or 200 mg plus a 100 mg injection on day 11 after birth. Pigs were weaned (~21 d of age) and allotted to nursery pens based on BW and corresponding treatment in a completely randomized design. During lactation, increasing injectable Fe up to 100 mg improved (quadratic; P < 0.05) average daily gain (ADG) and day 21 BW with no further improvement thereafter. There was no evidence of differences (P > 0.10) observed between the 200 mg and 200 mg + 100 mg treatments for growth. For the nursery period, increasing Fe dosage increased (linear; P < 0.05) ADG, average daily feed intake, and day 42 BW. There was no evidence of differences (P > 0.10) between the 200 mg and 200 mg + 100 mg treatments for nursery growth. For blood criteria, significant treatment × day interactions (P = 0.001) were observed for hemoglobin (Hb) and hematocrit (Hct). The interactions occurred because pigs that had <150 mg of injectable Fe had decreased values to day 21 and then increased to day 63 while pigs with 150 or 200 mg of injectable Fe had increased values to day 21 then stayed relatively constant to day 63. In summary, piglet performance during lactation was maximized at 100 mg while nursery growth performance and blood Fe status were maximized with a 200 mg Fe injection at processing. Providing an additional 100 mg of Fe on day 11 of age increased Hb, and Hct values at weaning and 14 d into the nursery but did not provide a growth performance benefit in lactation or nursery. These results indicate that providing 200 mg of injectable Fe provided from GleptoForte is sufficient to optimize lactation and subsequent nursery growth performance and blood Fe status.
Collapse
Affiliation(s)
- Hayden E Williams
- Department of Animal Sciences and Industry, College of Agriculture, Kansas State University, Manhattan, KS
| | - Joel M DeRouchey
- Department of Animal Sciences and Industry, College of Agriculture, Kansas State University, Manhattan, KS
| | - Jason C Woodworth
- Department of Animal Sciences and Industry, College of Agriculture, Kansas State University, Manhattan, KS
| | - Steven S Dritz
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS
| | - Michael D Tokach
- Department of Animal Sciences and Industry, College of Agriculture, Kansas State University, Manhattan, KS
| | - Robert D Goodband
- Department of Animal Sciences and Industry, College of Agriculture, Kansas State University, Manhattan, KS
| | | | - Eduarda M Bortoluzzi
- Department of Animal Sciences and Industry, College of Agriculture, Kansas State University, Manhattan, KS
| | - Jordan T Gebhardt
- Department of Animal Sciences and Industry, College of Agriculture, Kansas State University, Manhattan, KS
| |
Collapse
|
33
|
Effects of Iron Deficiency on Serum Metabolome, Hepatic Histology, and Function in Neonatal Piglets. Animals (Basel) 2020; 10:ani10081353. [PMID: 32764239 PMCID: PMC7460156 DOI: 10.3390/ani10081353] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 01/18/2023] Open
Abstract
Simple Summary Iron deficiency is a serious nutrient deficiency in neonatal pigs during the suckling period in modern intensive farming systems and leads to impaired immune response, infection risks, and retardation of growth. The objective was to determine how iron deficiency in neonatal pigs alters the serum metabolomic profile using quantitative and qualitative analysis by ultra-performance liquid chromatography-tandem mass spectrometer (UPLCMS/MS). The current results revealed that iron deficiency led to a series of metabolic changes involved in tyrosine metabolism, phenylalanine metabolism, bile secretion, primary bile acid biosynthesis, steroid biosynthesis, and upregulated activities of the urea cycle enzymes in the liver of neonatal piglets. Abstract Few studies focused on the effects of iron on characterizing alterations of metabolic processes in neonatal piglets. In the present study, 16 neonatal piglets were randomly assigned to two groups. In the first group piglets were given an intramuscularly injection of iron dextran at 150 mg as a positive control (CON) and the second group were not supplemented with iron as a negative control for iron deficiency (ID). At day 8, iron status, serum biochemical parameters, serum metabolome, hepatic histology, and hepatic expression of genes for the metabolism were analyzed. Results indicated that piglets without iron supplementation had significantly reduced iron values and increased blood urea nitrogen concentrations at day 8 (p < 0.05). Analysis of serum metabolome revealed that concentrations of serum lysine, leucine, tyrosine, methionine, and cholesterol were significantly decreased while concentrations of 3-Methyldioxyindole, chenodeoxycholate acid, indoleacetic acid, icosadienoic acid, phenylpyruvic acid, pantothenic acid, ursocholic acid, and cholic acid were significantly increased in iron deficient piglets (p < 0.05). Furthermore, expressions of cyp7a1 and the urea cycle enzyme (ornithinetranscarbamoylase and argininosuccinate synthetase) were significantly increased in iron deficient pigs (p < 0.05). The present experimental results indicated that neonatal piglets without iron supplementation drop to borderline anemia within 8 days after birth. Iron deficiency led to a series of metabolic changes involved in tyrosine metabolism, phenylalanine metabolism, bile secretion, primary bile acid biosynthesis, steroid biosynthesis, and upregulated activities of the urea cycle enzymes in the liver of neonatal piglets, suggesting early effects on metabolic health of neonatal piglets.
Collapse
|
34
|
Ding H, Yu X, Chen L, Han J, Zhao Y, Feng J. Tolerable upper intake level of iron damages the intestine and alters the intestinal flora in weaned piglets. Metallomics 2020; 12:1356-1369. [DOI: 10.1039/d0mt00096e] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Iron is an indispensable element for animal growth but become toxic at high concentrations, while tolerable upper intake level of iron shows adverse effect in the intestine.
Collapse
Affiliation(s)
- Haoxuan Ding
- College of Animal Science
- Zhejiang University
- Key Laboratory of animal feed and nutrition of Zhejiang Province
- Hangzhou
- China
| | - Xiaonan Yu
- College of Animal Science
- Zhejiang University
- Key Laboratory of animal feed and nutrition of Zhejiang Province
- Hangzhou
- China
| | - Lingjun Chen
- College of Animal Science
- Zhejiang University
- Key Laboratory of animal feed and nutrition of Zhejiang Province
- Hangzhou
- China
| | - Jianan Han
- College of Animal Science
- Zhejiang University
- Key Laboratory of animal feed and nutrition of Zhejiang Province
- Hangzhou
- China
| | - Yang Zhao
- College of Animal Science
- Zhejiang University
- Key Laboratory of animal feed and nutrition of Zhejiang Province
- Hangzhou
- China
| | - Jie Feng
- College of Animal Science
- Zhejiang University
- Key Laboratory of animal feed and nutrition of Zhejiang Province
- Hangzhou
- China
| |
Collapse
|
35
|
Abstract
Intestinal iron homeostasis is like the Zhong-Yong in traditional Chinese culture, which is a dynamic balance between Yin and Yang.
Collapse
Affiliation(s)
- Haoxuan Ding
- College of Animal Science
- Zhejiang University
- Key Laboratory of animal feed and nutrition of Zhejiang Province
- Hangzhou
- China
| | - Xiaonan Yu
- College of Animal Science
- Zhejiang University
- Key Laboratory of animal feed and nutrition of Zhejiang Province
- Hangzhou
- China
| | - Jie Feng
- College of Animal Science
- Zhejiang University
- Key Laboratory of animal feed and nutrition of Zhejiang Province
- Hangzhou
- China
| |
Collapse
|
36
|
Shannon MC, Hill GM. Trace Mineral Supplementation for the Intestinal Health of Young Monogastric Animals. Front Vet Sci 2019; 6:73. [PMID: 30918894 PMCID: PMC6424858 DOI: 10.3389/fvets.2019.00073] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 02/19/2019] [Indexed: 01/21/2023] Open
Abstract
Growth performance and feed efficiency are essential parameters when evaluating profitability of livestock. However, animal performance does not always reflect optimal gut health. Decades of research have supported the theory that improved animal performance such as average daily gain and feed efficiency can be impacted by intestinal health or the ability of the intestinal mucosa to absorb nutrients, but dysfunction may be found when the animal is stressed. Most of the early research focused on enteric infections causing diarrhea and nutritional alternatives to antibiotics which has led to findings related to pharmacological supplementation of trace minerals above the nutrient requirements for non-ruminants. While pharmacological concentrations of copper (Cu) have been shown to enhance growth, the mechanism in the gut is elusive. High concentrations of zinc (Zn) fed to newly weaned nursery pigs reduced the incidence of diarrhea from the proliferation of enterotoxigenic Escherichia coli (E. coli) and Clostridium and improve gut morphology. There are numerous publications where pharmacological supplementation of Zn as zinc oxide (ZnO) were fed to newly weaned pigs. Pharmacological Zn has been reported to shape the intestinal microflora as well as the diversity of the microflora during the first 2 weeks post-weaning. Both Fe deficiency and fortification impact bacterial growth in the intestine. Therefore, this paper will focus on the role of trace minerals that potentially impact optimal gut health of young monogastric animals.
Collapse
Affiliation(s)
| | - Gretchen Myers Hill
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
37
|
Zhuo Z, Yu X, Li S, Fang S, Feng J. Heme and Non-heme Iron on Growth Performances, Blood Parameters, Tissue Mineral Concentration, and Intestinal Morphology of Weanling Pigs. Biol Trace Elem Res 2019; 187:411-417. [PMID: 29770950 DOI: 10.1007/s12011-018-1385-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/09/2018] [Indexed: 12/22/2022]
Abstract
This experiment was conducted to evaluate the effects of heme and non-heme iron sources on growth performances, blood parameters, tissue mineral concentration, and intestinal morphology in weanling pigs. At 25 days of age, 32 newly weaned piglets (Duroc × Landrace × Yorkshire; 8.66 ± 0.59 kg) were allocated to one of the following dietary treatments: control group (basal diet with no extra iron addition), FeSO4 group (basal diet + 100 mg Fe/kg as FeSO4), Fe-Gly group (basal diet + 100 mg Fe/kg as Fe-Gly), and Heme group (basal diet + 100 mg Fe/kg as Heme). Each treatment had eight replicates and one pig per replicate. The experiment lasted for 28 days. The results showed that compared with basal diet, supplement with 100 mg/kg iron can increase ADG of the piglets, especially in the late experiment period (15~28 days). Heme significantly increased the a* value of longissimus dorsi muscle of piglets when compared with other iron sources (P < 0.05). The iron supplementations had no significant effect on hematological parameters, while Fe-Gly and heme increased pigs' serum iron content on day 28 when compared with FeSO4 and basal diet (P < 0.05). The liver iron deposition in pigs fed Fe-Gly and heme was also higher than those fed FeSO4 or basal diet (P < 0.05). Besides, diet supplement with iron significantly increased villus height (P < 0.05) in duodenum and it had tendency to increase villus height and crypt depth ratio in duodenum (P = 0.095). In conclusion, iron supplementation in diets can improve piglet's body iron state and intestinal development, but Fe-Gly and heme exhibited better bioavailability than traditional additive of FeSO4.
Collapse
Affiliation(s)
- Zhao Zhuo
- Key Laboratory of Animal Nutrition & Feed Science, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Xiaonan Yu
- Key Laboratory of Animal Nutrition & Feed Science, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Sisi Li
- Key Laboratory of Animal Nutrition & Feed Science, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Shenglin Fang
- Key Laboratory of Animal Nutrition & Feed Science, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Jie Feng
- Key Laboratory of Animal Nutrition & Feed Science, College of Animal Science, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
38
|
El-Sheikh AA, Ameen SH, AbdEl-Fatah SS. Ameliorating Iron Overload in Intestinal Tissue of Adult Male Rats: Quercetin vs Deferoxamine. J Toxicol 2018; 2018:8023840. [PMID: 30584425 PMCID: PMC6280249 DOI: 10.1155/2018/8023840] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 10/03/2018] [Accepted: 10/09/2018] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE The aim of our study is to compare the role of the new natural alternative (Quercetin) with the current iron-chelation therapy (Deferoxamine (DFO)) in the effect of iron overload on small intestinal tissues and to investigate the possible underlying molecular mechanisms of such toxicity. METHODS Forty-two adult male albino rats were divided into six groups: control groups, DFO, Quercetin, iron overload, iron overload+DFO, and iron overload+Quercetin groups. Animals received daily intraperitoneal injection of Deferoxamine (125 mg /kg), Quercetin (10 mg/kg), and ferric dextran (200 mg/kg) for 2 weeks. RESULTS Iron overloaded group showed significant increase in serum iron, total iron binding capacity (TIBC), transferrin saturation percentage (TS %) hepcidin (HEPC), serum ferritin, nontransferrin bound iron (NTBI), and small intestinal tissues iron levels. Iron overload significantly increased the serum oxidative stress indicator (MDA) and reduced serum total antioxidant capacity (TAC). On the other hand, iron overload increased IL6 and reduced IL10 in small intestinal tissues reflecting inflammatory condition and increased caspase 3 reactivity indicating apoptosis and increased iNOs expressing cell indicting oxidative stress especially in ileum. In addition, it induced small intestinal tissues pathological alterations. The treatment with Quercetin showed nonsignificant differences as compared to treatment with DFO that chelated the serum and tissue iron and improved the oxidative stress and reduced tissue IL6 and increased IL10 and decreased caspase 3 and iNOs expressing cells in small intestinal tissues. Moreover, it ameliorated the iron overload induced pathological alterations. CONCLUSION Our study showed the potential role of Quercetin as iron chelator like DFO in case of iron overload induced small intestinal toxicity in adult rats because of its serum and tissue iron chelation, improvement of serum, and small intestinal oxidative stress, ameliorating iron induced intestinal inflammation, apoptosis, and histopathological alterations.
Collapse
Affiliation(s)
- Arwa A. El-Sheikh
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig University, Egypt
| | - Shimaa Hamed Ameen
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig University, Egypt
| | | |
Collapse
|
39
|
Cao S, Wu H, Wang C, Zhang Q, Jiao L, Lin F, Hu CH. Diquat-induced oxidative stress increases intestinal permeability, impairs mitochondrial function, and triggers mitophagy in piglets. J Anim Sci 2018; 96:1795-1805. [PMID: 29562342 DOI: 10.1093/jas/sky104] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/16/2018] [Indexed: 12/20/2022] Open
Abstract
In the present study, we investigated the influence of diquat-induced oxidative stress on intestinal barrier, mitochondrial function, and the level of mitophagy in piglets. Twelve male Duroc × Landrace × Yorkshire 35-d-old pigs (weaned at 21 d of age), with an average body of 9.6 kg, were allotted to two treatments of six piglets each including the challenged group and the control group. The challenged pigs were injected with 100 mg/kg bodyweight diquat and control pigs injected with 0.9% (w/v) NaCl solution. The results showed that diquat injection decreased ADFI and ADG. Diquat decreased (P < 0.05) the activities of superoxide dismutase and glutathione peroxidase and increased (P < 0.05) the malondialdehyde concentrations. The lower (P < 0.05) transepithelial electrical resistance and higher (P < 0.05) paracellular permeability of fluorescein isothiocyanatedextran 4 kDa were found in diquat challenged piglets. Meanwhile, diquat decreased (P < 0.05) the protein abundance of claudin-1, occluding, and zonula occludens-1 in jejunum compared with the control group. Diquat-induced mitochondrial dysfunction, as demonstrated by increased (P < 0.05) reactive oxygen species production and decreased (P < 0.05) membrane potential of intestinal mitochondria. Diquat-injected pigs revealed a decrease (P < 0.05) of mRNA abundance of genes related to mitochondrial biogenesis and functions, PPARg coactivator-1α, mammalian-silencing information regulator-1, nuclear respiratory factor-1, mt transcription factor A, mt single-strand DNA-binding protein, mt polymerase r, glucokinase, citrate synthase, ATP synthase, and cytochrome coxidase subunit I and V in the jejunum. Diquat induced an increase (P < 0.05) in expression of mitophagy-related proteins, phosphatase and tensin homologue deleted on chromosome 10-induced putative kinase, and Parkin in the intestinal mitochondria, as well as an enhancement of the ratio of light chain 3-II (LC3-II) to LC3-I content in the jejunal mucosa. These results suggest that oxidative stress disrupted the intestinal barrier, caused mitochondrial dysfunction, and triggered mitophagy.
Collapse
Affiliation(s)
- Shuting Cao
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Huan Wu
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - ChunChun Wang
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Qianhui Zhang
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Lefei Jiao
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Fanghui Lin
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Caihong H Hu
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| |
Collapse
|
40
|
Wang W, Van Noten N, Degroote J, Romeo A, Vermeir P, Michiels J. Effect of zinc oxide sources and dosages on gut microbiota and integrity of weaned piglets. J Anim Physiol Anim Nutr (Berl) 2018; 103:231-241. [PMID: 30298533 DOI: 10.1111/jpn.12999] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 09/01/2018] [Accepted: 09/03/2018] [Indexed: 12/17/2022]
Abstract
Zinc oxide (ZnO) supplied at pharmacological dosage in diets of weaned piglets improves growth performance. However, it causes environmental contamination and induces bacterial antibiotic resistance, yet this practice is debated. The effects on gut microbiota and integrity in weaned piglets of conventional ZnO at nutritional and pharmacological dosage (110 and 2,400 mg/kg Zn, respectively) were compared to an alternative ZnO source at 110 and 220 mg/kg Zn. Each of the four treatments was applied to four pens (two piglets/pen; weaning age, 20 days) for 15 days, and piglets were sampled on day 15 to determine indices of gut integrity. Feeding conventional ZnO at 2,400 mg/kg Zn reduced coliforms and Escherichia coli in distal small intestine as compared to conventional ZnO at 110 mg/kg (-1.7 and -1.4 log10 cfu/g, respectively), whereas the alternative ZnO reduced only coliforms, irrespective of dosage (-1.6 to -1.7 log10 cfu/g). Transepithelial electrical resistance of distal small intestinal mucosa was higher for pigs fed the alternative ZnO source as compared with groups fed 110 mg/kg Zn of conventional ZnO, in line with a trend for higher gene expression of claudin-1 and zona occludens-1. Interestingly, the alternative ZnO source at 110 and 220 mg/kg Zn increased intestinal alkaline phosphatase gene transcript as compared to conventional ZnO at 110 mg/kg Zn, whereas the alternative ZnO source at 110 mg/kg Zn exhibited higher Zn concentrations in mucosa (2,520 μg/g) as compared to conventional ZnO at 110 mg/kg Zn (1,211 μg/g). However, assessing alkaline phosphatase activity, no significant effects were found. In conclusion, the alternative ZnO reduced digesta Enterobacteriaceae numbers and improved gut integrity, albeit similar or better, depending on the dosage, to the effects of pharmacological dosage of conventional ZnO.
Collapse
Affiliation(s)
- Wei Wang
- Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium
| | - Noémie Van Noten
- Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium
| | - Jeroen Degroote
- Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium
| | | | - Pieter Vermeir
- Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium
| | - Joris Michiels
- Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium
| |
Collapse
|
41
|
Cao ST, Wang CC, Wu H, Zhang QH, Jiao LF, Hu CH. Weaning disrupts intestinal antioxidant status, impairs intestinal barrier and mitochondrial function, and triggers mitophagy in piglets. J Anim Sci 2018; 96:1073-1083. [PMID: 29617867 DOI: 10.1093/jas/skx062] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In the present study, we investigated the influence of weaning on antioxidant status, intestinal integrity, mitochondrial function, and the mitophagy level in piglets (weaned at 21 d) during the 1 wk after weaning. The redox status was measured by antioxidant enzymes activities, related genes expression, and malondialdehyde (MDA) content in jejunum. The intestinal barrier function was assessed by the Ussing chamber and expression of tight junction proteins in the jejunum. The function of intestine mitochondria was measured by mitochondrial DNA (mtDNA) content and activities of mitochondria oxidative phosphorylation complexes. The levels of light chain 3-1 (LC3-I), light chain 3-II (LC3-II), PTEN-induced putative kinase 1 (PINK1), and Parkin were determined to investigate whether mitophagy is involved in the weaning process. The results showed that, as compared with the preweaning phase (d 0), weaning suppressed (P < 0.05) the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) on d 3 and d 7 postweaning, decreased (P < 0.05) the expression of copper and zinc superoxide dismutase (Cu/Zn-SOD), manganese-containing superoxide dismutase (Mn-SOD) on d 3 postweaning, declined (P < 0.05) the level of glutathione peroxidase 1 (GPX-1) and glutathione peroxidase 4 (GPX-4) on d 3 and d 7 postweaning, and increased (P < 0.05) MDA content in jejunum on d 3 and d 7 postweaning. The jejunal transepithelial electrical resistance and levels of occludin, claudin-1, and zonula occludens-1 on d 3 and d 7 postweaning were reduced (P < 0.05), and paracellular flux of fluorescein isothiocyanatedextran (4 kDa) on d 3 and d 7 postweaning was increased (P < 0.05). Weaning induced mitochondrial dysfunction, as demonstrated by decreased (P < 0.05) content of mtDNA on d 3 and d 7 postweaning and declined (P < 0.05) activities of mitochondria complexes (I, II, III, IV) in jejunum on d 1, d 3, and d 7 postweaning. Weaning led to an increased (P < 0.05) expression level of mitophagy-related proteins, PINK1 and Parkin, in the intestinal mitochondria, as well as an enhancement (P < 0.05) of the ratio of LC3-II to LC3-I content in the jejunal mucosa on d 1, d 3, and d 7 postweaning. These results suggest that weaning disrupted intestinal oxidative balance, and this imbalance may impair intestinal barrier and mitochondrial function and trigger mitophagy in piglets.
Collapse
Affiliation(s)
- S T Cao
- Animal Science College, Zhejiang University, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - C C Wang
- Animal Science College, Zhejiang University, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - H Wu
- Animal Science College, Zhejiang University, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Q H Zhang
- Animal Science College, Zhejiang University, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - L F Jiao
- Animal Science College, Zhejiang University, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - C H Hu
- Animal Science College, Zhejiang University, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| |
Collapse
|
42
|
Chronic social stress in pigs impairs intestinal barrier and nutrient transporter function, and alters neuro-immune mediator and receptor expression. PLoS One 2017; 12:e0171617. [PMID: 28170426 PMCID: PMC5295718 DOI: 10.1371/journal.pone.0171617] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/23/2017] [Indexed: 12/17/2022] Open
Abstract
Psychosocial stress is a major factor driving gastrointestinal (GI) pathophysiology and disease susceptibility in humans and animals. The mechanisms governing susceptibility to stress-induced GI disease remain poorly understood. In the present study, we investigated the influence of chronic social stress (CSS) in pigs, induced by 7 d of chronic mixing/crowding stress, on intestinal barrier and nutrient transport function, corticotropin releasing factor (CRF) signaling and immunological responses. Results from this study showed that CSS resulted in a significant impairment of ileal and colonic barrier function indicated by reduced transepithelial electrical resistance (TER) in the ileum and increased FD4 flux in the ileum (by 0.8 fold) and colon (by 0.7 fold). Ileal sodium glucose linked transporter 1 (SGLT-1) function, measured as glucose-induced changes in short-circuit current (Isc), was diminished (by 52%) in CSS pigs, associated with reduced body weight gain and feed efficiency. Although reductions in SGLT-1 function were observed in CSS pigs, mRNA expression for SGLT-1, villus heights were increased in CSS pigs. Corticotropin releasing factor (CRF) mRNA was upregulated (by 0.9 fold) in the ileum of CSS pigs but not in the colon. Urocortin 2 (Ucn2) mRNA was upregulated (by 1.5 fold) in the colon of CSS pigs, but not in the ileum. In CSS pigs, a downregulation of pro-inflammatory cytokines mRNA (IL1B, TNFA, IL8, and IL6) was observed in both ileum and colon, compared with controls. In contrast CSS induced a marked upregulation of mRNA for IL10 and mast cell chymase gene (CMA1) in the ileum and colon. Together, these data demonstrate that chronic stress in pigs results in significant alterations in intestinal barrier and nutrient transport function and neuro-immune mediator and receptor expression.
Collapse
|
43
|
Paganini D, Uyoga MA, Zimmermann MB. Iron Fortification of Foods for Infants and Children in Low-Income Countries: Effects on the Gut Microbiome, Gut Inflammation, and Diarrhea. Nutrients 2016; 8:nu8080494. [PMID: 27529276 PMCID: PMC4997407 DOI: 10.3390/nu8080494] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 07/25/2016] [Accepted: 08/01/2016] [Indexed: 12/19/2022] Open
Abstract
Iron deficiency anemia (IDA) is common among infants and children in Sub-Saharan Africa and is a leading contributor to the global burden of disease, as well as a hindrance to national development. In-home iron fortification of complementary foods using micronutrient powders (MNPs) effectively reduces the risk for IDA by ensuring that the iron needs of infants and young children are met without changing their traditional diet. However, the iron dose delivered by MNPs is high, and comparable on a mg iron per kg body weight to the supplemental doses (2 mg/kg) typically given to older children, which increases diarrhea risk. In controlled studies, iron-containing MNPs modestly increase risk for diarrhea in infants; in some cases, the diarrhea is severe and may require hospitalization. Recent in vitro and in vivo studies provide insights into the mechanism of this effect. Provision of iron fortificants to school-age children and iron-containing MNPs to weaning infants decreases the number of beneficial ‘barrier’ commensal gut bacteria (e.g., bifidobacteria), increases the enterobacteria to bifidobacteria ratio and abundances of opportunistic pathogens (e.g., pathogenic Escherichia coli), and induces gut inflammation. Thus, although iron-containing MNPs are highly effective in reducing IDA, they may increase gastrointestinal morbidity in infants, and safer formulations are needed.
Collapse
Affiliation(s)
- Daniela Paganini
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, ETH Zurich, Zurich 8092, Switzerland.
| | - Mary A Uyoga
- College of Health Sciences, Jomo Kenyatta University of Agriculture and Technology, Nairobi 00200, Kenya.
| | - Michael B Zimmermann
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, ETH Zurich, Zurich 8092, Switzerland.
| |
Collapse
|