1
|
Sonálio KC, Malcorra de Almeida L, Bassi LS, Kuritza LN, Dias IDC, da Rocha C, Maiorka A. Effect of Putrescine Inoculation In Ovo on Hatchability, Hepatic and Muscular Glycogen Reserve, Intestinal Morphology, and Performance of Broilers. Animals (Basel) 2025; 15:1259. [PMID: 40362073 PMCID: PMC12070924 DOI: 10.3390/ani15091259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/22/2025] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
The objective of this study was to evaluate the inoculation of nutrient solutions with increasing levels of putrescine on the hatchability, physiology, and performance of broilers during the initial phase. The study is composed of four treatments with increasing doses of putrescine (0.015; 0.030; 0.060, and 0.090%) and a control group. At hatch, hatchability rate; ratio between egg weight and chick weight; ratio between yolk sac, liver, breast and intestine weight, and chick weight; glycogen concentration in the liver and breast; and morphometric characteristics of the jejunum and ileum were evaluated. After hatch, 400 birds were housed in metabolic cages according to the treatments received, and feed intake, body weight gain, and feed conversion ratio were assessed at specific time points. Hatchability, chick performance at hatch, and organ weight were not affected by the inoculation of increasing levels of putrescine. Intestinal villi at hatch were higher in groups supplemented with putrescine (p < 0.001). The amounts of hepatic glycogen per gram of tissue at hatch were higher in groups with the lowest levels of putrescine and decreased with increasing doses (p = 0.017). Growth performance from 1 to 21 days was not influenced by the inoculation of putrescine.
Collapse
Affiliation(s)
- Katiucia Cristine Sonálio
- Department of Veterinary Science, Federal University of Paraná, Curitiba 80035-050, PR, Brazil; (L.M.d.A.); (L.N.K.); (I.d.C.D.); (A.M.)
| | - Leopoldo Malcorra de Almeida
- Department of Veterinary Science, Federal University of Paraná, Curitiba 80035-050, PR, Brazil; (L.M.d.A.); (L.N.K.); (I.d.C.D.); (A.M.)
| | - Lucas Schmidt Bassi
- Department of Animal Science, Federal University of Paraná, Curitiba 80035-050, PR, Brazil (C.d.R.)
| | - Leandro Nagae Kuritza
- Department of Veterinary Science, Federal University of Paraná, Curitiba 80035-050, PR, Brazil; (L.M.d.A.); (L.N.K.); (I.d.C.D.); (A.M.)
| | - Isabela de Camargo Dias
- Department of Veterinary Science, Federal University of Paraná, Curitiba 80035-050, PR, Brazil; (L.M.d.A.); (L.N.K.); (I.d.C.D.); (A.M.)
| | - Chayane da Rocha
- Department of Animal Science, Federal University of Paraná, Curitiba 80035-050, PR, Brazil (C.d.R.)
| | - Alex Maiorka
- Department of Veterinary Science, Federal University of Paraná, Curitiba 80035-050, PR, Brazil; (L.M.d.A.); (L.N.K.); (I.d.C.D.); (A.M.)
| |
Collapse
|
2
|
Zhang C, Zhen Y, Weng Y, Lin J, Xu X, Ma J, Zhong Y, Wang M. Research progress on the microbial metabolism and transport of polyamines and their roles in animal gut homeostasis. J Anim Sci Biotechnol 2025; 16:57. [PMID: 40234982 PMCID: PMC11998418 DOI: 10.1186/s40104-025-01193-x] [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: 12/03/2024] [Accepted: 03/09/2025] [Indexed: 04/17/2025] Open
Abstract
Polyamines (putrescine, spermidine, and spermine) are aliphatic compounds ubiquitous in prokaryotes and eukaryotes. Positively charged polyamines bind to negatively charged macromolecules, such as nucleic acids and acidic phospholipids, and are involved in physiological activities including cell proliferation, differentiation, apoptosis and gene regulation. Intracellular polyamine levels are regulated by biosynthesis, catabolism and transport. Polyamines in the body originate from two primary sources: dietary intake and intestinal microbial metabolism. These polyamines are then transported into the bloodstream, through which they are distributed to various tissues and organs to exert their biological functions. Polyamines synthesized by intestinal microorganisms serve dual critical roles. First, they are essential for maintaining polyamine concentrations within the digestive tract. Second, through transcriptional and post-transcriptional mechanisms, these microbial-derived polyamines modulate the expression of genes governing key processes in intestinal epithelial cells-including proliferation, migration, apoptosis, and cell-cell interactions. Collectively, these regulatory effects help maintain intestinal epithelial homeostasis and ensure the integrity of the gut barrier. In addition, polyamines interact with the gut microbiota to maintain intestinal homeostasis by promoting microbial growth, biofilm formation, swarming, and endocytosis vesicle production, etc. Supplementation with polyamines has been demonstrated to be important in regulating host intestinal microbial composition, enhancing nutrient absorption, and improving metabolism and immunity. In this review, we will focus on recent advances in the study of polyamine metabolism and transport in intestinal microbes and intestinal epithelial cells. We then summarize the scientific understanding of their roles in intestinal homeostasis, exploring the advances in cellular and molecular mechanisms of polyamines and their potential clinical applications, and providing a rationale for polyamine metabolism as an important target for the treatment of intestinal-based diseases.
Collapse
Affiliation(s)
- Chong Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Yongkang Zhen
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Yunan Weng
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Jiaqi Lin
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Xinru Xu
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Jianjun Ma
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Yuhong Zhong
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Sciences, Shihezi, 832000, China.
| |
Collapse
|
3
|
Yu C, Li H, Hua L, Che L, Feng B, Fang Z, Xu S, Zhuo Y, Li J, Wu D, Zhang J, Lin Y. Deciphering the microbiome, lipopolysaccharides, and metabolome interplay: Unveiling putrescine's mechanism for enhancing sperm quality in heat-stressed boars. Theriogenology 2025; 236:60-73. [PMID: 39919573 DOI: 10.1016/j.theriogenology.2025.01.027] [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: 05/16/2024] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 02/09/2025]
Abstract
Global warming has added to concerns regarding declining male fertility due to high temperatures. As a metabolite of arginine, putrescine improves gut health and promotes testicular development in boar; however, its action in improving semen quality under heat stress is unknown. Therefore, we assessed the effect of putrescine on the semen quality of boars in a heat stress model. Results showed that putrescine ameliorated the heat stress-induced decline in semen quality and testosterone levels in boars, confirmed by sperm viability, immobility rate, and apoptosis levels. Fecal microbial 16S rRNA sequencing showed that heat stress induces intestinal microecological dysregulation triggering an increase in the serum lipopolysaccharide (LPS) levels and reducing boar semen quality. A negative correlation between the Lachnospiraceae_XPB1014_group and LPS-binding protein (LBP) levels was observed. The Lachnospiraceae_XPB1014_group was reduced significantly under heat stress, and its relative abundance significantly increased after putrescine diet, which reduced both LPS and LBP in the serum of heat-stressed boars. Heat stress also affected plasma amino acid metabolism, and the regulation of plasma metabolism by putrescine can be attributed to its effects on LPS and the LBP owing to the significantly correlation of both with multiple plasma differential metabolites. Putrescine is thus considered to inhibit the increased serum LPS by acting on intestinal microorganisms, particularly by increasing the relative abundance of the Lachnospiraceae_XPB1014_group, and further modulate plasma amino acid metabolism to improve the semen quality in heat-stressed boars.
Collapse
Affiliation(s)
- Chenglong Yu
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Hao Li
- Tieqi Lishi Food Co., Ltd, Guizhou, China College of Life Science, Sichuan Agricultural University, Ya'an, 625014, Sichuan, China
| | - Lun Hua
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Lianqiang Che
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Bin Feng
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Zhengfeng Fang
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Shengyu Xu
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yong Zhuo
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Jian Li
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - De Wu
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Junjie Zhang
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, Sichuan, China
| | - Yan Lin
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed Science, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
4
|
Rahman R, Fouhse JM, Ju T, Fan Y, Bhardwaj T, Brook RK, Nosach R, Harding J, Willing BP. The impact of wild-boar-derived microbiota transplantation on piglet microbiota, metabolite profile, and gut proinflammatory cytokine production differs from sow-derived microbiota. Appl Environ Microbiol 2025; 91:e0226524. [PMID: 39902926 PMCID: PMC11921332 DOI: 10.1128/aem.02265-24] [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: 11/13/2024] [Accepted: 12/19/2024] [Indexed: 02/06/2025] Open
Abstract
Colonization of co-evolved, species-specific microbes in early life plays a crucial role in gastrointestinal development and immune function. This study hypothesized that modern pig production practices have resulted in the loss of co-evolved species and critical symbiotic host-microbe interactions. To test this, we reintroduced microbes from wild boars (WB) into conventional piglets to explore their colonization dynamics and effects on gut microbial communities, metabolite profiles, and immune responses. At postnatal day (PND) 21, 48 piglets were assigned to four treatment groups: (i) WB-derived mixed microbial community (MMC), (ii) sow-derived MMC, (iii) a combination of WB and sow MMC (Mix), or (iv) Control (PBS). Post-transplantation analyses at PND 48 revealed distinct microbial communities in WB-inoculated piglets compared with Controls, with trends toward differentiation from Sow but not Mix groups. WB-derived microbes were more successful in colonizing piglets, particularly in the Mix group, where they competed with Sow-derived microbes. WB group cecal digesta enriched with Lactobacillus helveticus, Lactobacillus mucosae, and Lactobacillus pontis. Cecal metabolite analysis showed that WB piglets were enriched in histamine, acetyl-ornithine, ornithine, citrulline, and other metabolites, with higher histamine levels linked to Lactobacillus abundance. WB piglets exhibited lower cecal IL-1β and IL-6 levels compared with Control and Sow groups, whereas the Mix group showed reduced IFN-γ, IL-2, and IL-6 compared with the Sow group. No differences in weight gain, fecal scores, or plasma cytokines were observed, indicating no adverse effects. These findings support that missing WB microbes effectively colonize domestic piglets and may positively impact metabolite production and immune responses.IMPORTANCEThis study addresses the growing concern over losing co-evolved, species-specific microbes in modern agricultural practices, particularly in pig production. The implementation of strict biosecurity measures and widespread antibiotic use in conventional farming systems may disrupt crucial host-microbe interactions that are essential for gastrointestinal development and immune function. Our research demonstrates that by reintroducing wild boar-derived microbes into domestic piglets, these microbes can successfully colonize the gut, influence microbial community composition, and alter metabolite profiles and immune responses without causing adverse effects. These findings also suggest that these native microbes can fill an intestinal niche, positively impacting immune activation. This research lays the groundwork for future strategies to enhance livestock health and performance by restoring natural microbial populations that produce immune-modulating metabolites.
Collapse
Affiliation(s)
- Rajibur Rahman
- Department of Agricultural Food & Nutritional Science, Faculty of Agricultural, Life & Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Janelle M. Fouhse
- Department of Agricultural Food & Nutritional Science, Faculty of Agricultural, Life & Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Tingting Ju
- Department of Agricultural Food & Nutritional Science, Faculty of Agricultural, Life & Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Yi Fan
- Department of Agricultural Food & Nutritional Science, Faculty of Agricultural, Life & Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Tulika Bhardwaj
- Department of Agricultural Food & Nutritional Science, Faculty of Agricultural, Life & Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada
- University of Calgary, Calgary, Alberta, Canada
| | - Ryan K. Brook
- College of Agriculture and Bioresources, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Roman Nosach
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - John Harding
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Benjamin P. Willing
- Department of Agricultural Food & Nutritional Science, Faculty of Agricultural, Life & Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
5
|
Fan S, Wang W, Che W, Xu Y, Jin C, Dong L, Xia Q. Nanomedicines Targeting Metabolic Pathways in the Tumor Microenvironment: Future Perspectives and the Role of AI. Metabolites 2025; 15:201. [PMID: 40137165 PMCID: PMC11943624 DOI: 10.3390/metabo15030201] [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: 01/30/2025] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Tumor cells engage in continuous self-replication by utilizing a large number of resources and capabilities, typically within an aberrant metabolic regulatory network to meet their own demands. This metabolic dysregulation leads to the formation of the tumor microenvironment (TME) in most solid tumors. Nanomedicines, due to their unique physicochemical properties, can achieve passive targeting in certain solid tumors through the enhanced permeability and retention (EPR) effect, or active targeting through deliberate design optimization, resulting in accumulation within the TME. The use of nanomedicines to target critical metabolic pathways in tumors holds significant promise. However, the design of nanomedicines requires the careful selection of relevant drugs and materials, taking into account multiple factors. The traditional trial-and-error process is relatively inefficient. Artificial intelligence (AI) can integrate big data to evaluate the accumulation and delivery efficiency of nanomedicines, thereby assisting in the design of nanodrugs. Methods: We have conducted a detailed review of key papers from databases, such as ScienceDirect, Scopus, Wiley, Web of Science, and PubMed, focusing on tumor metabolic reprogramming, the mechanisms of action of nanomedicines, the development of nanomedicines targeting tumor metabolism, and the application of AI in empowering nanomedicines. We have integrated the relevant content to present the current status of research on nanomedicines targeting tumor metabolism and potential future directions in this field. Results: Nanomedicines possess excellent TME targeting properties, which can be utilized to disrupt key metabolic pathways in tumor cells, including glycolysis, lipid metabolism, amino acid metabolism, and nucleotide metabolism. This disruption leads to the selective killing of tumor cells and disturbance of the TME. Extensive research has demonstrated that AI-driven methodologies have revolutionized nanomedicine development, while concurrently enabling the precise identification of critical molecular regulators involved in oncogenic metabolic reprogramming pathways, thereby catalyzing transformative innovations in targeted cancer therapeutics. Conclusions: The development of nanomedicines targeting tumor metabolic pathways holds great promise. Additionally, AI will accelerate the discovery of metabolism-related targets, empower the design and optimization of nanomedicines, and help minimize their toxicity, thereby providing a new paradigm for future nanomedicine development.
Collapse
Affiliation(s)
| | | | | | | | | | - Lei Dong
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (S.F.); (W.W.); (W.C.); (Y.X.); (C.J.)
| | - Qin Xia
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (S.F.); (W.W.); (W.C.); (Y.X.); (C.J.)
| |
Collapse
|
6
|
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
|
7
|
Wang R, Yu H, Li A, Wang T, Wang Q, Qi H, Wang C, Zhang J. Maslinic acid prevented lipopolysaccharide-induced injury of IPEC-J2 cells through regulating PTEN-FAK signaling pathway. J Cell Physiol 2024; 239:e31446. [PMID: 39311471 DOI: 10.1002/jcp.31446] [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: 03/15/2024] [Revised: 09/05/2024] [Accepted: 09/13/2024] [Indexed: 12/18/2024]
Abstract
Intestinal epithelial injury is one of the typical symptoms associated with intestinal inflammation and diarrhea, and the repair of the intestinal epithelium intricately linked to cell migration. Here, we test the hypothesis that maslinic acid (MA) regulates porcine intestinal epithelial cell migration by inhibiting focal adhesion kinase (FAK)/AKT signaling pathway. In this experiment, the optimal concentration of MA (0.5 μg/mL) on IPEC-J2 cell viability was selected to investigate the effect under low-dose lipopolysaccharide (LPS) (1 μg/mL) conditions. Transcriptome sequencing and polymerase chain reaction array results revealed that MA could alleviate LPS-induced the gene expressions decreasing in focal adhesion signaling pathway. From the pathway map analysis and western blot analysis results, MA alleviated the LPS-induced decrease in FAK protein expression mainly by promoting FAK protein phosphorylation, which in turn alleviated the decrease in cell migration and formation of cytoskeleton protein Vinculin and F-actin, the above results were verified by FAK phosphorylation inhibitors Defactinib. The molecular docking and immunoprecipitation further verified that MA could bind to PTEN protein and significantly inhibit its interaction with FAK protein, blocking the function of PTEN to inhibit FAK phosphorylation finally shown to promote the level of FAK phosphorylation, meanwhile LPS inhibited FAK protein expression and its binding to PKC and PTEN proteins. Our study revealed the role of MA and LPS in FAK protein, and increased understanding of MA anti-inflammatory mechanism.
Collapse
Affiliation(s)
- Rui Wang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Hao Yu
- College of Animal Sciences, Jilin University, Changchun, China
| | - Aike Li
- Academy of National Food and Strategic Reserves Administration, Beijing, China
| | - Ting Wang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Qiyuan Wang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Huiyu Qi
- College of Animal Sciences, Jilin University, Changchun, China
| | - Chuanqi Wang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Jing Zhang
- College of Animal Sciences, Jilin University, Changchun, China
- Academy of National Food and Strategic Reserves Administration, Beijing, China
| |
Collapse
|
8
|
Ma WW, Huang ZQ, Liu K, Li DZ, Mo TL, Liu Q. The role of intestinal microbiota and metabolites in intestinal inflammation. Microbiol Res 2024; 288:127838. [PMID: 39153466 DOI: 10.1016/j.micres.2024.127838] [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: 01/18/2024] [Revised: 06/11/2024] [Accepted: 07/13/2024] [Indexed: 08/19/2024]
Abstract
With the imbalance of intestinal microbiota, the body will then face an inflammatory response, which has serious implications for human health. Bodily allergies, injury or pathogens infections can trigger or promote inflammation and alter the intestinal environment. Meanwhile, excessive changes in the intestinal environment cause the imbalance of microbial homeostasis, which leads to the proliferation and colonization of opportunistic pathogens, invasion of the body's immune system, and the intensification of inflammation. Some natural compounds and gut microbiota and metabolites can reduce inflammation; however, the details of how they interact with the gut immune system and reduce the gut inflammatory response still need to be fully understood. The review focuses on inflammation and intestinal microbiota imbalance caused by pathogens. The body reacts differently to different types of pathogenic bacteria, and the ingestion of pathogens leads to inflamed gastrointestinal tract disorders or intestinal inflammation. In this paper, unraveling the interactions between the inflammation, pathogenic bacteria, and intestinal microbiota based on inflammation caused by several common pathogens. Finally, we summarize the effects of intestinal metabolites and natural anti-inflammatory substances on inflammation to provide help for related research of intestinal inflammation caused by pathogenic bacteria.
Collapse
Affiliation(s)
- Wen-Wen Ma
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China
| | - Zhi-Qiang Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China
| | - Kun Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China
| | - De-Zhi Li
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China
| | - Tian-Lu Mo
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China.
| | - Qing Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China.
| |
Collapse
|
9
|
Barreiro-Alonso E, Castro-Estrada P, Sánchez M, Peña-Iglesias P, Suárez L, Cantabrana B. Association of Polyamine Intake, Other Dietary Components, and Fecal Content of N-acetyl Putrescine and Cadaverine with Patients' Colorectal Lesions. Nutrients 2024; 16:2894. [PMID: 39275210 PMCID: PMC11397480 DOI: 10.3390/nu16172894] [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: 07/10/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/16/2024] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer death worldwide. Early detection and the modification of risk factors, such as diet, can reduce its incidence. Among food components, polyamines are important for maintaining gastrointestinal health and are metabolites of gut microbiota. Their disruption is linked to CRC, making polyamines a potential marker of the disease. This study analyzed the relationship between dietary components, including polyamines, and the presence of polyamines in feces to determine whether their presence could contribute to predicting the occurrence of colorectal lesions in patients. In total, 59 participants of both sexes (aged 50 to 70 years) who had undergone colonoscopy screening for CRC (18 without and 41 with colorectal lesions) participated in the study. A nutritional survey and determination of fecal polyamine content were performed. Specific dietary components and putrescine levels were higher in patients with colorectal lesions. The diet ratio of putrescine-spermidine and the fecal content of N-acetyl putrescine and cadaverine were elevated in patients with precancerous lesions and adenocarcinomas, showing a potential predictive value for the presence of colorectal lesions. These findings suggest that N-acetyl putrescine and cadaverine could be complementary markers for the diagnosis of suspected colorectal lesions.
Collapse
Affiliation(s)
- Eva Barreiro-Alonso
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Servicio de Digestivo, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain
| | - Paula Castro-Estrada
- Farmacología, Departamento de Medicina, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Manuel Sánchez
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Farmacología, Departamento de Medicina, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Pablo Peña-Iglesias
- Farmacología, Departamento de Medicina, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Lorena Suárez
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Farmacología, Departamento de Medicina, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Begoña Cantabrana
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Farmacología, Departamento de Medicina, Universidad de Oviedo, 33006 Oviedo, Spain
| |
Collapse
|
10
|
Zhou M, Wu Z, Deng D, Wang B, Zhou X, Zhou B, Wang C, Zeng Y. Effects of taurine on the growth performance, diarrhea, oxidative stress and intestinal barrier function of weanling piglets. Front Vet Sci 2024; 11:1436282. [PMID: 39170630 PMCID: PMC11336868 DOI: 10.3389/fvets.2024.1436282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 07/29/2024] [Indexed: 08/23/2024] Open
Abstract
Oxidative damage resulting from weaning stress significantly impacts the growth performance and health status of piglets. Taurine, a dietary antioxidant with diverse functions, was investigated in this study for its protective role against weaning stress-induced oxidative damage and its underlying mechanism. Forty 28-day-old male castrated weaned piglets were randomly assigned to four groups. The control group received the basal diet, while the experimental groups were fed the basal diet supplemented with 0.1, 0.2%, or 0.3% taurine over a 28-day period. In vitro, H2O2 was utilized to induce oxidative damage to the jejunal mucosa of piglets via IPEC-J2 cells. The results demonstrated that taurine supplementation reduced the incidence of diarrhea in piglets compared to that in the control group (p < 0.05); the addition of 0.2 and 0.3% taurine led to increased average daily gain and improved feed conversion efficiency in weaned piglets, showing a linear dose-response correlation (p < 0.05). Taurine supplementation at 0.2 and 0.3% enhanced the activities of serum CAT and GSH-Px while decreasing the levels of serum NO, XOD, GSSG, and MDA (p < 0.05). Moreover, it significantly elevated the levels of GSS, Trx, POD, complex I, mt-nd5, and mt-nd6, enhancing superoxide anion scavenging capacity and the hydroxyl-free scavenging rate in the livers of weaned piglets while reducing NO levels in the liver (p < 0.05). Additionally, 0.2 and 0.3% taurine supplementation decreased serum IL-6 levels and elevated the concentrations of IgA, IgG, and IL-10 in weaned piglets (p < 0.05). The levels of occludin, claudin, and ZO-1 in the jejunum mucosa of weaned piglets increased with 0.2 and 0.3% taurine supplementation (p < 0.05). In IPEC-J2 cells, pretreatment with 25 mM taurine for 24 h enhanced the activities of SOD and CAT; reduced the MDA content; upregulated the mRNA expression of various genes, including ZO-1, occludin, claudin-1, Nrf2, and HO-1; and reversed the oxidative damage induced by H2O2 exposure (p < 0.05). Overall, the findings suggest that the inclusion of 2 and 3% taurine in the diet can enhance growth performance, reduce diarrhea rates, ameliorate oxidative stress and inflammation, and promote intestinal barrier function in weaned piglets.
Collapse
Affiliation(s)
- Miao Zhou
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Zichen Wu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Donghua Deng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Bin Wang
- Hunan Institute of Microbiology, Changsha, China
| | | | - Bingyu Zhou
- Hunan Institute of Microbiology, Changsha, China
| | | | - Yan Zeng
- Hunan Institute of Microbiology, Changsha, China
| |
Collapse
|
11
|
Oliero M, Cuisiniere T, Ajayi AS, Gerkins C, Hajjar R, Fragoso G, Calvé A, Vennin Rendos H, Mathieu-Denoncourt A, Dagbert F, De Broux É, Loungnarath R, Schwenter F, Sebajang H, Ratelle R, Wassef R, Richard C, Duperthuy M, Gravel AE, Vincent AT, Santos MM. Putrescine Supplementation Limits the Expansion of pks+ Escherichia coli and Tumor Development in the Colon. CANCER RESEARCH COMMUNICATIONS 2024; 4:1777-1792. [PMID: 38934090 PMCID: PMC11261243 DOI: 10.1158/2767-9764.crc-23-0355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 04/11/2024] [Accepted: 06/24/2024] [Indexed: 06/28/2024]
Abstract
Escherichia coli that harbor the polyketide synthase (pks) genomic island produce colibactin and are associated with sporadic colorectal cancer development. Given the considerable prevalence of pks+ bacteria in healthy individuals, we sought to identify strategies to limit the growth and expansion of pks+ E. coli. We found that culture supernatants of the probiotic strain E. coli Nissle 1917 were able to inhibit the growth of the murine pathogenic strain pks+ E. coli NC101 (EcNC101). We performed a nontargeted analysis of the metabolome in supernatants from several E. coli strains and identified putrescine as a potential postbiotic capable of suppressing EcNC101 growth in vitro. The effect of putrescine supplementation was then evaluated in the azoxymethane/dextran sulfate sodium mouse model of colorectal cancer in mice colonized with EcNC101. Putrescine supplementation inhibited the growth of pks+ E. coli, reduced the number and size of colonic tumors, and downmodulated the release of inflammatory cytokines in the colonic lumen. Additionally, putrescine supplementation led to shifts in the composition and function of gut microbiota, characterized by an increase in the Firmicutes/Bacteroidetes ratio and enhanced acetate production. The effect of putrescine was further confirmed in vitro using a pks+ E. coli strain isolated from a patient with colorectal cancer. These results suggest that probiotic-derived metabolites can be used as an alternative to live bacteria in individuals at risk of developing colorectal cancer due to the presence of pks+ bacteria in their colon. SIGNIFICANCE Putrescine supplementation inhibits the growth of cancer-promoting bacteria in the gut, lowers inflammation, and reduces colon cancer development. The consumption of healthy foods rich in putrescine may be a potential prophylactic approach for individuals at risk of developing colorectal cancer due to the presence of pks+ bacteria in their colon.
Collapse
Affiliation(s)
- Manon Oliero
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
| | - Thibault Cuisiniere
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
| | - Ayodeji S. Ajayi
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
| | - Claire Gerkins
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
| | - Roy Hajjar
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montréal, Canada.
| | - Gabriela Fragoso
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
| | - Annie Calvé
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
| | - Hervé Vennin Rendos
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
| | - Annabelle Mathieu-Denoncourt
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montréal, Canada.
| | - François Dagbert
- Digestive Surgery Service, Department of Surgery, Centre hospitalier de l’Université de Montréal (CHUM), Montréal, Canada.
| | - Éric De Broux
- Digestive Surgery Service, Department of Surgery, Centre hospitalier de l’Université de Montréal (CHUM), Montréal, Canada.
| | - Rasmy Loungnarath
- Digestive Surgery Service, Department of Surgery, Centre hospitalier de l’Université de Montréal (CHUM), Montréal, Canada.
| | - Frank Schwenter
- Digestive Surgery Service, Department of Surgery, Centre hospitalier de l’Université de Montréal (CHUM), Montréal, Canada.
| | - Herawaty Sebajang
- Digestive Surgery Service, Department of Surgery, Centre hospitalier de l’Université de Montréal (CHUM), Montréal, Canada.
| | - Richard Ratelle
- Digestive Surgery Service, Department of Surgery, Centre hospitalier de l’Université de Montréal (CHUM), Montréal, Canada.
| | - Ramses Wassef
- Digestive Surgery Service, Department of Surgery, Centre hospitalier de l’Université de Montréal (CHUM), Montréal, Canada.
| | - Carole Richard
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montréal, Canada.
- Digestive Surgery Service, Department of Surgery, Centre hospitalier de l’Université de Montréal (CHUM), Montréal, Canada.
| | - Marylise Duperthuy
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montréal, Canada.
| | - Andrée E. Gravel
- Drug Discovery Platform, Research Institute McGill University Health Centre, Montreal, Canada.
| | - Antony T. Vincent
- Département des sciences animales, Faculté des sciences de l’agriculture et de l’alimentation, Université Laval, Quebec City, Canada.
- Institut de biologie intégrative et des systèmes, Université Laval, Quebec City, Canada.
| | - Manuela M. Santos
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada.
| |
Collapse
|
12
|
Gázquez A, García-Serna AM, Hernández-Caselles T, Martín-Orozco E, Cantero-Cano E, Prieto-Sánchez MT, Molina-Ruano MD, Castillo-Lacalle R, Vioque J, Morales E, García-Marcos L, Larqué E. Plasma polyamines during pregnancy and their relationships with maternal allergies and the immune response of the neonates. Pediatr Allergy Immunol 2024; 35:e14167. [PMID: 38860435 DOI: 10.1111/pai.14167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/22/2024] [Accepted: 05/13/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Some studies have reported that polyamine levels may influence immune system programming. The aim of this study was to evaluate the polyamine profile during gestation and its associations with maternal allergy and cytokine production in cord blood cells in response to different allergenic stimuli. METHODS Polyamines were determined in plasma of pregnant women (24 weeks, N = 674) and in umbilical cord samples (N = 353 vein and N = 160 artery) from the Mediterranean NELA birth cohort. Immune cell populations were quantified, and the production of cytokines in response to different allergic and mitogenic stimuli was assessed in cord blood. RESULTS Spermidine and spermine were the most prevalent polyamines in maternal, cord venous, and cord arterial plasma. Maternal allergies, especially allergic conjunctivitis, were associated with lower spermine in umbilical cord vein. Higher levels of polyamines were associated with higher lymphocyte number but lower Th2-related cells in cord venous blood. Those subjects with higher levels of circulating polyamines in cord showed lower production of inflammatory cytokines, especially IFN-α, and lower production of Th2-related cytokines, mainly IL-4 and IL-5. The effects of polyamines on Th1-related cytokines production were uncertain. CONCLUSIONS Spermidine and spermine are the predominant polyamines in plasma of pregnant women at mid-pregnancy and also in umbilical cord. Maternal allergic diseases like allergic conjunctivitis are related to lower levels of polyamines in cord vein, which could influence the immune response of the newborn. Cord polyamine content is related to a decreased Th2 response and inflammatory cytokines production, which might be important to reduce an allergenic phenotype in the neonate.
Collapse
Affiliation(s)
- Antonio Gázquez
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
- Department of Physiology, University of Murcia, Murcia, Spain
| | - Azahara M García-Serna
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
- Department of Public Health Sciences, University of Murcia, Murcia, Spain
| | - Trinidad Hernández-Caselles
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
- Department of Biochemistry and Molecular Biology B and Immunology, Faculty of Medicine, University of Murcia, Murcia, Spain
- Network of Asthma and Adverse and Allergic Reactions (ARADyAL), Madrid, Spain
| | - Elena Martín-Orozco
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
- Department of Biochemistry and Molecular Biology B and Immunology, Faculty of Medicine, University of Murcia, Murcia, Spain
- Network of Asthma and Adverse and Allergic Reactions (ARADyAL), Madrid, Spain
| | | | - María T Prieto-Sánchez
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
- Obstetrics & Gynaecology Service, "Virgen de la Arrixaca" University Clinical Hospital, University of Murcia, Murcia, Spain
| | - María D Molina-Ruano
- Obstetrics & Gynaecology Service, "Virgen de la Arrixaca" University Clinical Hospital, University of Murcia, Murcia, Spain
| | - Rafaela Castillo-Lacalle
- Obstetrics & Gynaecology Service, "Virgen de la Arrixaca" University Clinical Hospital, University of Murcia, Murcia, Spain
| | - Jesús Vioque
- Health and Biomedical Research Institute of Alicante (ISABIAL-UMH), Alicante, Spain
- CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Eva Morales
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
- Department of Public Health Sciences, University of Murcia, Murcia, Spain
- CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Luís García-Marcos
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
- Network of Asthma and Adverse and Allergic Reactions (ARADyAL), Madrid, Spain
| | - Elvira Larqué
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
- Department of Physiology, University of Murcia, Murcia, Spain
| |
Collapse
|
13
|
Moore ML, Ford JL, Schladweiler MC, Dye JA, Jackson TW, Miller CN. Gut metabolic changes during pregnancy reveal the importance of gastrointestinal region in sample collection. Metabolomics 2024; 20:40. [PMID: 38460019 PMCID: PMC11168590 DOI: 10.1007/s11306-024-02099-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/06/2024] [Indexed: 03/11/2024]
Abstract
INTRODUCTION Studies of gastrointestinal physiology and the gut microbiome often consider the influence of intestinal region on experimental endpoints. However, this same consideration is not often applied to the gut metabolome. Understanding the contribution of gut regionality may be critically important to the rapidly changing metabolic environments, such as during pregnancy. OBJECTIVES We sought to characterize the difference in the gut metabolome in pregnant mice stratified by region-comparing the small intestine, cecum, and feces. Pre-pregnancy feces were collected to understand the influence of pregnancy on the fecal metabolome. METHODS Feces were collected from CD-1 female mice before breeding. On gestation day (GD) 18, gut contents were collected from the small intestine, cecum, and descending colon. Metabolites were analyzed with LC-MS/MS using the Biocrates MetaboINDICATOR™ MxP® Quant 500 kit. RESULTS Of the 104 small molecule metabolites meeting analysis criteria, we found that 84 (81%) were differentially abundant based on gut region. The most significant regional comparison observed was between the cecum and small intestines, with 52 (50%) differentially abundant metabolites. Pregnancy itself altered 41 (39.4%) fecal small molecule metabolites. CONCLUSIONS The regional variation observed in the gut metabolome are likely due to the microbial and physiological differences between the different parts of the intestines. Additionally, pregnancy impacts the fecal metabolome, which may be due to evolving needs of both the dam and fetus.
Collapse
Affiliation(s)
- Makala L Moore
- Oak Ridge Institute for Science and Education, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Jermaine L Ford
- Chemical Characterization and Exposure Division, Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Mette C Schladweiler
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Janice A Dye
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Thomas W Jackson
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA.
| | - Colette N Miller
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA.
| |
Collapse
|
14
|
Hu C, Ji F, Lv R, Zhou H, Hou G, Xu T. Putrescine promotes MMP9-induced angiogenesis in skeletal muscle through hydrogen peroxide/METTL3 pathway. Free Radic Biol Med 2024; 212:433-447. [PMID: 38159892 DOI: 10.1016/j.freeradbiomed.2023.12.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/25/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
Blood vessels play a crucial role in the development of skeletal muscle, ensuring the supply of nutrients and oxygen. Putrescine, an essential polyamine for eukaryotic cells, has an unclear impact on skeletal muscle angiogenesis. In this study, we observed lower vessel density and reduced putrescine level in the muscle of low-birth-weight piglet models, and identified a positive correlation between putrescine content and muscle vessel density. Furthermore, putrescine was found to promote angiogenesis in skeletal muscle both in vitro and in vivo by targeting matrix metalloproteinase 9 (MMP9). On a mechanistic level, putrescine augmented the expression of methyltransferase like 3 (METTL3) by attenuating hydrogen peroxide production, thereby increasing the level of N6-methyladenosine (m6A)-modified MMP9 mRNA. This m6A-modified MMP9 mRNA was subsequently recognized and bound by the YTH N6-methyladenosine RNA binding protein 1 (YTHDF1), enhancing the stability of MMP9 mRNA and its protein expression, consequently accelerating angiogenesis in skeletal muscle. In summary, our findings suggest that putrescine enhances MMP9-mediated angiogenesis in skeletal muscle via the hydrogen peroxide/METTL3 pathway.
Collapse
Affiliation(s)
- Chengjun Hu
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China; Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, 524013, China
| | - Fengjie Ji
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China
| | - Renlong Lv
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China
| | - Hanlin Zhou
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China; Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, 524013, China
| | - Guanyu Hou
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China.
| | - Tieshan Xu
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China.
| |
Collapse
|
15
|
Calabrese E, Hayes AW, Pressman P, Kapoor R, Dhawan G, Calabrese V, Agathokleous E. Polyamines and hormesis: Making sense of a dose response dichotomy. Chem Biol Interact 2023; 386:110748. [PMID: 37816449 DOI: 10.1016/j.cbi.2023.110748] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/08/2023] [Accepted: 09/22/2023] [Indexed: 10/12/2023]
Abstract
The diverse biological effects of polyamines (putrescine, spermidine and spermine) were reviewed in the context of hormesis in an integrative manner for the first time. The findings illustrate that each of these polyamines commonly induces hormetic dose responses in a wide range of biological models and types of cells for multiple endpoints in numerous plant species and animal models. Plant research emphasized preconditioning experimental studies in which the respective polyamines conferred some protection against the damaging effects of a broad range of environmental stressors such as drought, salinity, cold/heat, heavy metals and UV-damage in an hormetic manner. Polyamine-based animal hormesis studies emphasized biomedical endpoints such as longevity and neuroprotection. These findings have important biological and biomedical implications and should guide experimental designs of low dose investigations.
Collapse
Affiliation(s)
- Edward Calabrese
- Department of Environmental Health Sciences, Morrill I, N344, University of Massachusetts, Amherst, MA, 01003, USA.
| | - A Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, FL, USA.
| | - Peter Pressman
- University of Maine, 5728 Fernald Hall, Room 201, Orono, ME, 04469, USA.
| | - Rachna Kapoor
- Saint Francis Hospital and Medical Center, Hartford, CT, USA.
| | | | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, School of Medicine University of Catania, Via Santa Sofia 97, Catania, 95123, Italy.
| | - Evgenios Agathokleous
- School of Applied Meteorology, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| |
Collapse
|
16
|
Shan J, Liu S, Liu H, Yuan J, Lin J. Mechanism of Qingchang Suppository on repairing the intestinal mucosal barrier in ulcerative colitis. Front Pharmacol 2023; 14:1221849. [PMID: 37675045 PMCID: PMC10478270 DOI: 10.3389/fphar.2023.1221849] [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: 05/13/2023] [Accepted: 08/07/2023] [Indexed: 09/08/2023] Open
Abstract
Ulcerative colitis (UC) is a refractory inflammatory bowel disease, and the outcomes of conventional therapies of UC, including 5-aminosalicylic acid, glucocorticoids, immunosuppressants, and biological agents, are not satisfied with patients and physicians with regard to adverse reactions and financial burden. The abnormality of the intestinal mucosal barrier in the pathogenesis of UC was verified. Qingchang Suppository (QCS) is an herbal preparation and is effective in treating ulcerative proctitis. The mechanism of QCS and its active ingredients have not been concluded especially in mucosal healing. This review elucidated the potential mechanism of QCS from the intestinal mucosal barrier perspective to help exploring future QCS research directions.
Collapse
Affiliation(s)
- Jingyi Shan
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Suxian Liu
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Haoyue Liu
- Department of Intensive Care Unit, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianye Yuan
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiang Lin
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
17
|
Hodkovicova N, Halas S, Tosnerova K, Stastny K, Svoboda M. The use of functional amino acids in different categories of pigs - A review. VET MED-CZECH 2023; 68:299-312. [PMID: 37982122 PMCID: PMC10646542 DOI: 10.17221/72/2023-vetmed] [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: 06/29/2023] [Accepted: 08/11/2023] [Indexed: 11/21/2023] Open
Abstract
The present review deals with a particularly important topic: the use of functional amino acids in different categories of pigs. It is especially relevant in the context of the current efforts to reduce the use of antibiotics in pig farming and the search for possible alternatives to replace them. The review is based on the definition that functional amino acids (FAAs) are classified as dispensable amino acids, but with additional biological functions, i.e., not only are they used for protein formation, but they are also involved in regulating essential metabolic pathways to improve health, survival, growth, and development. We describe the mechanism of action of individual FAAs and their potential use in pigs, including glutamate, glutamine, arginine, branched-chain amino acids (i.e., leucine, isoleucine, and valine), tryptophan and glycine. The work is divided into three parts. The first part deals with the FAAs and their role in the overall health of sows and their offspring. The second part describes the use of functional amino acids in piglets after weaning. Part three examines the use of functional amino acids in growing and fattening pigs and their impact on meat quality.
Collapse
Affiliation(s)
- Nikola Hodkovicova
- Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Brno, Czech Republic
| | - Simon Halas
- Department of Animal Nutrition and Husbandry, University of Veterinary Medicine and Pharmacy in Kosice, Kosice, Slovak Republic
| | - Kristina Tosnerova
- Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Brno, Czech Republic
| | - Kamil Stastny
- Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Brno, Czech Republic
| | - Martin Svoboda
- Ruminant and Swine Clinic, University of Veterinary Sciences Brno, Brno, Czech Republic
| |
Collapse
|
18
|
Vasquez R, Kim SH, Oh JK, Song JH, Hwang IC, Kim IH, Kang DK. Multispecies probiotic supplementation in diet with reduced crude protein levels altered the composition and function of gut microbiome and restored microbiome-derived metabolites in growing pigs. Front Microbiol 2023; 14:1192249. [PMID: 37485501 PMCID: PMC10360209 DOI: 10.3389/fmicb.2023.1192249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/23/2023] [Indexed: 07/25/2023] Open
Abstract
Both crude protein (CP) and probiotics can modulate the gut microbiome of the host, thus conferring beneficial effects. However, the benefits of low CP diet supplemented with multispecies probiotics on gut microbiome and its metabolites have not been investigated in pigs. Thus, we investigated the combinatory effects of low CP diet supplemented with multispecies probiotics on gut microbiome composition, function, and microbial metabolites in growing pigs. In total, 140 6 week-old piglets (Landrace × Yorkshire × Duroc) were used in this study. The pigs were divided into four groups with a 2 × 2 factorial design based on their diets: normal-level protein diet (16% CP; NP), low-level protein diet (14% CP; LP), NP with multispecies probiotics (NP-P), and LP with multispecies probiotics (LP-P). After the feeding trial, the fecal samples of the pigs were analyzed. The fecal scores were improved by the probiotic supplementation, especially in LP-P group. We also observed a probiotic-mediated alteration in the gut microbiome of pigs. In addition, LP-P group showed higher species richness and diversity compared with other groups. The addition of multispecies probiotics in low CP diet also enhanced gut microbiota metabolites production, such as short-chain fatty acids (SCFAs) and polyamines. Correlation analysis revealed that Oscillospiraceae UCG-002, Eubacterium coprostanoligenes, Lachnospiraceae NK4A136 group, and Muribaculaceae were positively associated with SCFAs; and Prevotella, Eubacterium ruminantium, Catenibacterium, Alloprevotella, Prevotellaceae NK3B31 group, Roseburia, Butyrivibrio, and Dialister were positively correlated with polyamines. Supplementation with multispecies probiotics modulated the function of the gut microbiome by upregulating the pathways for protein digestion and utilization, potentially contributing to enriched metabolite production in the gut. The results of this study demonstrate that supplementation with multispecies probiotics may complement the beneficial effects of low CP levels in pig feed. These findings may help formulate sustainable feeding strategies for swine production.
Collapse
|
19
|
Panah FM, Lauridsen C, Højberg O, Jensen HE, Nielsen TS. Composition of mucus- and digesta-associated bacteria in growing pigs with and without diarrhea differed according to the presence of colonic inflammation. BMC Microbiol 2023; 23:145. [PMID: 37210480 DOI: 10.1186/s12866-023-02874-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/28/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND In the pig production, diarrhea can occur during different growth stages including the period 4-16 weeks post weaning, during which a diarrheal outbreak also termed as colitis-complex diarrhea (CCD) can occur and it is distinct from post-weaning diarrhea (1-2 weeks post weaning). We hypothesized that CCD in growing pigs is associated with changes in colonic microbiota composition and fermentation patterns, and the aim of the present observational study was to identify changes in digesta-associated bacteria (DAB) and mucus-associated bacteria (MAB) in the colon of growing pigs with and without diarrhea. A total number of 30 pigs (8, 11, and 12 weeks of age) were selected; 20 showed clinical signs of diarrhea and 10 appeared healthy. Based on histopathological examination of colonic tissues, 21 pigs were selected for further studies and classified as follows: without diarrhea, no colon inflammation (NoDiar; n = 5), with diarrhea, without colonic inflammation (DiarNoInfl; n = 4), and with diarrhea, with colonic inflammation (DiarInfl; n = 12). Composition (based on 16S rRNA gene amplicon sequencing) and fermentation pattern (short-chain fatty acids; SCFA profile) of the DAB and MAB communities were characterized. RESULTS The DAB showed higher alpha diversity compared to MAB in all pigs, and both DAB and MAB showed lowest alpha diversity in the DiarNoInfl group. Beta diversity was significantly different between DAB and MAB as well as between diarrheal groups in both DAB and MAB. Compared to NoDiar, DiarInfl showed increased abundance of various taxa, incl. certain pathogens, in both digesta and mucus, as well as decreased digesta butyrate concentration. However, DiarNoInfl showed reduced abundance of different genera (mainly Firmicutes) compared to NoDiar, but still lower butyrate concentration. CONCLUSION Diversity and composition of MAB and DAB changed in diarrheal groups depending on presence/absence of colonic inflammation. We also suggest that DiarNoInfl group was at the earlier stage of diarrhea compared with DiarInfl, with a link to dysbiosis of colonic bacterial composition as well as reduced butyrate concentration, which plays a pivotal role in gut health. This could have led to diarrhea with inflammation due to a dysbiosis, associated with an increase in e.g., Escherichia-Shigella (Proteobacteria), Helicobacter (Campylobacterota), and Bifidobacterium (Actinobacteriota), which may tolerate or utilize oxygen and cause epithelial hypoxia and inflammation. The increased consumption of oxygen in epithelial mucosal layer by infiltrated neutrophils may also have added up to this hypoxia. Overall, the results confirmed that changes in DAB and MAB were associated with CCD and reduced butyrate concentration in digesta. Moreover, DAB might suffice for future community-based studies of CCD.
Collapse
Affiliation(s)
- Farhad M Panah
- Department of Animal and Veterinary Sciences, Aarhus University, Tjele, Denmark
| | - Charlotte Lauridsen
- Department of Animal and Veterinary Sciences, Aarhus University, Tjele, Denmark
| | - Ole Højberg
- Department of Animal and Veterinary Sciences, Aarhus University, Tjele, Denmark.
| | - Henrik Elvang Jensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tina Skau Nielsen
- Department of Animal and Veterinary Sciences, Aarhus University, Tjele, Denmark
| |
Collapse
|
20
|
Liu B, Paudel S, Flowers WL, Piedrahita JA, Wang X. Uterine histotroph and conceptus development: III. Adrenomedullin stimulates proliferation, migration and adhesion of porcine trophectoderm cells via AKT-TSC2-MTOR cell signaling pathway. Amino Acids 2023:10.1007/s00726-023-03265-6. [PMID: 37036518 DOI: 10.1007/s00726-023-03265-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 03/29/2023] [Indexed: 04/11/2023]
Abstract
Adrenomedullin (ADM) as a highly conserved peptide hormone has been reported to increase significantly in the uterine lumen during the peri-implantation period of pregnancy in pigs, but its functional roles in growth and development of porcine conceptus (embryonic/fetus and its extra-embryonic membranes) as well as underlying mechanisms remain largely unknown. Therefore, we conducted in vitro experiments using our established porcine trophectoderm cell line (pTr2) isolated from Day-12 porcine conceptuses to test the hypothesis that porcine ADM stimulates cell proliferation, migration and adhesion via activation of mechanistic target of rapamycin (MTOR) cell signaling pathway in pTr2 cells. Porcine ADM at 10-7 M stimulated (P < 0.05) pTr2 cell proliferation, migration and adhesion by 1.4-, 1.5- and 1.2-folds, respectively. These ADM-induced effects were abrogated (P < 0.05) by siRNA-mediated knockdown of ADM (siADM) and its shared receptor component calcitonin-receptor-like receptor (CALCRL; siCALCRL), as well as by rapamycin, the inhibitor of MTOR. Using siRNA-mediated knockdown of CALCRL coupled with Western blot analyses, ADM signaling transduction was determined in which ADM binds to CALCRL to increase phosphorylation of MTOR, its downstream effectors (4EBP1, P70S6K, and S6), and upstream regulators (AKT and TSC2). Collectively, these results suggest that porcine ADM in histotroph acts on its receptor component CALCRL to activate AKT-TSC2-MTOR, particularly MTORC1 signaling cascade, leading to elongation, migration and attachment of conceptuses.
Collapse
Affiliation(s)
- Bangmin Liu
- Department of Animal Science, North Carolina State University, NC, 27695, Raleigh, USA
| | - Sudikshya Paudel
- Department of Animal Science, North Carolina State University, NC, 27695, Raleigh, USA
| | - William L Flowers
- Department of Animal Science, North Carolina State University, NC, 27695, Raleigh, USA
| | - Jorge A Piedrahita
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27695, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27695, USA
| | - Xiaoqiu Wang
- Department of Animal Science, North Carolina State University, NC, 27695, Raleigh, USA.
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27695, USA.
| |
Collapse
|
21
|
Sohn J, Li L, Zhang L, Genco RJ, Falkner KL, Tettelin H, Rowsam AM, Smiraglia DJ, Novak JM, Diaz PI, Sun Y, Kirkwood KL. Periodontal disease is associated with increased gut colonization of pathogenic Haemophilus parainfluenzae in patients with Crohn's disease. Cell Rep 2023; 42:112120. [PMID: 36774550 PMCID: PMC10415533 DOI: 10.1016/j.celrep.2023.112120] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 11/19/2022] [Accepted: 01/30/2023] [Indexed: 02/13/2023] Open
Abstract
Intestinal colonization of the oral bacterium Haemophilus parainfluenzae has been associated with Crohn's disease (CD) severity and progression. This study examines the role of periodontal disease (PD) as a modifier for colonization of H. parainfluenzae in patients with CD and explores the mechanisms behind H. parainfluenzae-mediated intestinal inflammation. Fifty subjects with and without CD were evaluated for the presence of PD, and their oral and fecal microbiomes were characterized. PD is associated with increased levels of H. parainfluenzae strains in subjects with CD. Oral inoculation of H. parainfluenzae elicits strain-dependent intestinal inflammation in murine models of inflammatory bowel disease, which is associated with increased intestinal interferon-γ (IFN-γ)+ CD4+ T cells and disruption of the host hypusination pathway. In summary, this study establishes a strain-specific pathogenic role of H. parainfluenzae in intestinal inflammation and highlights the potential effect of PD on intestinal colonization by pathogenic H. parainfluenzae strains in patients with CD.
Collapse
Affiliation(s)
- Jiho Sohn
- Department of Medicine, State University of New York at Buffalo, University at Buffalo, 645 Biomedical Research Building, 3435 Main Street, Buffalo, NY 14214, USA; Department of Oral Biology, State University of New York at Buffalo, University at Buffalo, Buffalo, NY 14214, USA.
| | - Lu Li
- Department of Oral Biology, State University of New York at Buffalo, University at Buffalo, Buffalo, NY 14214, USA
| | - Lixia Zhang
- Department of Oral Biology, State University of New York at Buffalo, University at Buffalo, Buffalo, NY 14214, USA
| | - Robert J Genco
- Department of Oral Biology, State University of New York at Buffalo, University at Buffalo, Buffalo, NY 14214, USA
| | - Karen L Falkner
- Department of Oral Biology, State University of New York at Buffalo, University at Buffalo, Buffalo, NY 14214, USA
| | - Hervé Tettelin
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Aryn M Rowsam
- Department of Cell Stress Biology, Reconstructive Surgery Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Dominic J Smiraglia
- Department of Cell Stress Biology, Reconstructive Surgery Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Jan M Novak
- Department of Medicine, State University of New York at Buffalo, University at Buffalo, 645 Biomedical Research Building, 3435 Main Street, Buffalo, NY 14214, USA
| | - Patricia I Diaz
- Department of Oral Biology, State University of New York at Buffalo, University at Buffalo, Buffalo, NY 14214, USA
| | - Yijun Sun
- Department of Microbiology and Immunology, State University of New York at Buffalo, University at Buffalo, Buffalo, NY 14214, USA
| | - Keith L Kirkwood
- Department of Oral Biology, State University of New York at Buffalo, University at Buffalo, Buffalo, NY 14214, USA; Department of Head & Neck/Plastic & Reconstructive Surgery Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| |
Collapse
|
22
|
Aminoclay Nanoparticles Induce Anti-Inflammatory Dendritic Cells to Attenuate LPS-Elicited Pro-Inflammatory Immune Responses. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248743. [PMID: 36557876 PMCID: PMC9787634 DOI: 10.3390/molecules27248743] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/23/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
Although 3-aminopropyl functionalized magnesium phyllosilicate nanoparticles (hereafter aminoclay nanoparticles, ACNs) are well-known nanomaterials employed as drug carriers, their effects on immune cells remain unclear. To address this issue, we explored murine dendritic cells (DCs) as these cells belong to the innate arm of the immune system and function as antigen-presenting cells to elicit adaptive immune responses. We examined the in vitro effects of ACNs on DCs isolated from B6 mice. ACN treatment significantly down-regulated the expression of inflammasome-related markers, including NLRP3, caspase-1, and IL1β. The ACNs-induced anti-inflammatory DC phenotype was further confirmed by down-regulation of the AKT/mTOR/HIF1α signaling pathway. Such anti-inflammatory effects of ACNs on DCs occurred independently of DC subtypes. To document the effects of ACNs on DCs more clearly, we examined their anti-inflammatory effects on lipopolysaccharide (LPS)-activated DCs. As expected, excessive inflammatory responses (increased mitochondrial ROS and Th1-type cytokines such as IL12 and IL1β) of LPS-activated DCs were dramatically attenuated by ACN treatment. Furthermore, ACNs down-regulated IFNγ production by antigen-specific CD4+ T cells, which is consistent with a reduced inflammatory phenotype of DCs. Overall, our results provide support for employing ACNs as drug delivery materials with therapeutic potential to control inflammatory disorders.
Collapse
|
23
|
Mo W, Liu G, Wu C, Jia G, Zhao H, Chen X, Wang J. STIM1 promotes IPEC-J2 porcine epithelial cell restitution by TRPC1 signaling. Anim Biotechnol 2022; 33:1492-1503. [PMID: 33866928 DOI: 10.1080/10495398.2021.1910044] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Intestinal epithelial restitution is partly dependent on cell migration, which reseals superficial wounding after injury. Here, we tested the hypothesis that stromal interaction molecule 1(STIM1) regulates porcine intestinal epithelial cell migration by activating transient receptor potential canonical 1 (TRPC1) signaling. Results showed that the knockdown of STIM1 repressed cell migration after wounding, reduced the protein concentration of STIM1 and TRPC1, and decreased the inositol trisphosphate (IP3) content in IPEC-J2 cells (p < 0.05). However, overexpression of STIM1 obtained opposite results (p < 0.05). The inhibition of TRPC1 activity by treatment with SKF96365 in cells overexpressing wild-type and mutant STIM1 attenuated the STIM1 overexpression-induced increase of cell migration, STIM1, TRPC1 and IP3 (p < 0.05). In addition, polyamine depletion caused by α-difluoromethylornithine (DFMO) resulted in the decrease of above-mentioned parameters, and exogenous polyamine could attenuate the negative effects of DFMO on IPEC-J2 cells (p < 0.05). Moreover, the overexpression of STIM1 could rescue cell migration, the protein level of STIM1 and TRPC1, and IP3 content in polyamine-deficient IPEC-J2 cells (p < 0.05). These results indicated that STIM1 could enhance porcine intestinal epithelial cell migration via the TRPC1 signaling pathway. Inhibition of cell migration by polyamine depletion resulted from the reduction of STIM1 activity.
Collapse
Affiliation(s)
- Weiwei Mo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu, China
| | - Guangmang Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu, China
| | - Caimei Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu, China
| | - Gang Jia
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu, China
| | - Hua Zhao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu, China
| | - Xiaoling Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Chengdu, China
| | - Jing Wang
- Maize Research Institute, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
24
|
Pretorius L, Van Staden AD, Kellermann TA, Henning N, Smith C. Rooibos (Aspalathus linearis) alters secretome trace amine profile of probiotic and commensal microbes in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2022; 297:115548. [PMID: 35850312 DOI: 10.1016/j.jep.2022.115548] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGY RELEVANCE Aspalathus linearis (Burm.f.) R. Dahlgren (rooibos) tea is anecdotally renowned for its calming effect in the context of gastrointestinal discomfort, but little scientific support is available to elucidate potential mechanisms of action. Enhancement of dietary polyphenol content to improve gut health via prebiotic-like modulation of the gut microbiota has gained significant research interest. Given the known high polyphenol content of rooibos, rooibos tea may potentially exert a prebiotic effect in the gut to facilitate an improvement in chronic inflammatory gastrointestinal conditions. AIM OF THE STUDY This study aimed to determine the prebiotic or health-modulating potential of rooibos tea in terms of its effect on gut microbial growth and secretome trace amine composition, as well as to determine how differential rooibos processing alters this activity. METHODS Three rooibos preparations (green and fermented leave aqueous extracts, as well as a green leaf ethanol extract) were compared in terms of their phenolic composition (qTOF-LC/MS). Moreover, the effect of rooibos exposure on growth and secretome trace amine levels of probiotic and commensal microbes were assessed (LC/MS). In addition, given the known female bias prevalent for many gastrointestinal disorders, experiments were conducted in the absence and presence of estradiol. RESULTS Polyphenolic composition of rooibos was drastically reduced by fermentation. Aqueous extracts of both green and fermented rooibos improved microbial growth, although fermented rooibos had the most pronounced effect (p < 0.01). In terms of secretome trace amine profile, both aqueous extracts of rooibos seemed to facilitate increased putrescine secretion (p < 0.0001) and decreased tryptamine production (p < 0.0001). Estradiol seemed to suppress trace amine secretion by bacteria (Lactobacillus plantarum, Lactobacillus reuteri and Enterococcus mundtii) but increased it in yeast (Saccharomyces boulardii). CONCLUSION Rooibos altered gut probiotic and commensal microbial growth and secretome trace amine profiles in vitro, suggesting it has potential to modulate gut microbial composition and functionality as a prebiotic. Current data suggest that these effects are highly dependent on raw material processing. Finally, rooibos may be able to prevent estradiol-associated alterations in trace amine profile, which may have important implications for patient management in female-predominant gastrointestinal disorders.
Collapse
Affiliation(s)
- L Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, South Africa.
| | - A D Van Staden
- Department of Microbiology, Faculty of Science, Stellenbosch University, South Africa; Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa.
| | - T A Kellermann
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa.
| | - N Henning
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa.
| | - C Smith
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa.
| |
Collapse
|
25
|
Song ZY, Yuan D, Zhang SX. Role of the microbiome and its metabolites in ankylosing spondylitis. Front Immunol 2022; 13:1010572. [PMID: 36311749 PMCID: PMC9608452 DOI: 10.3389/fimmu.2022.1010572] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
Ankylosing spondylitis (AS), a chronic condition that commonly influences the spine and sacroiliac joints, usually progresses to stiffness and progressive functional limitation. Its fundamental etiology and pathogenesis are likely multifactorial and remain elusive. As environmental factors, gut microbiota performs critical functions in the pathogenesis of AS through various mechanisms, including interacting with genes, enhancing intestinal permeability, activating the gut mucosa immune system, and affecting the intestinal microbiota metabolites. This review provides an overview of recent advances in investigating gut microbiota in AS pathogenesis and discusses potential methods for future therapeutic intervention.
Collapse
Affiliation(s)
- Zi-Yi Song
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Duo Yuan
- Department of Gynecology and Obstetrics, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Sheng-Xiao Zhang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
26
|
Lian J, Liang Y, Zhang H, Lan M, Ye Z, Lin B, Qiu X, Zeng J. The role of polyamine metabolism in remodeling immune responses and blocking therapy within the tumor immune microenvironment. Front Immunol 2022; 13:912279. [PMID: 36119047 PMCID: PMC9479087 DOI: 10.3389/fimmu.2022.912279] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
The study of metabolism provides important information for understanding the biological basis of cancer cells and the defects of cancer treatment. Disorders of polyamine metabolism is a common metabolic change in cancer. With the deepening of understanding of polyamine metabolism, including molecular functions and changes in cancer, polyamine metabolism as a new anti-cancer strategy has become the focus of attention. There are many kinds of polyamine biosynthesis inhibitors and transport inhibitors, but not many drugs have been put into clinical application. Recent evidence shows that polyamine metabolism plays essential roles in remodeling the tumor immune microenvironment (TIME), particularly treatment of DFMO, an inhibitor of ODC, alters the immune cell population in the tumor microenvironment. Tumor immunosuppression is a major problem in cancer treatment. More and more studies have shown that the immunosuppressive effect of polyamines can help cancer cells to evade immune surveillance and promote tumor development and progression. Therefore, targeting polyamine metabolic pathways is expected to become a new avenue for immunotherapy for cancer.
Collapse
Affiliation(s)
- Jiachun Lian
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yanfang Liang
- Department of Pathology, Dongguan Hospital Affiliated to Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Hailiang Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Minsheng Lan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Ziyu Ye
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Department of Pathology, Dongguan Hospital Affiliated to Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
- Dongguan Metabolite Analysis Engineering Technology Center of Cells for Medical Use, Guangdong Xinghai Institute of Cell, Dongguan, China
| | - Bihua Lin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Key Laboratory of Medical Bioactive Molecular Research for Department of Education of Guangdong Province, Collaborative Innovation Center for Antitumor Active Substance Research and Development, Zhanjiang, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Zhanjiang, China
| | - Xianxiu Qiu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Key Laboratory of Medical Bioactive Molecular Research for Department of Education of Guangdong Province, Collaborative Innovation Center for Antitumor Active Substance Research and Development, Zhanjiang, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Zhanjiang, China
| | - Jincheng Zeng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Dongguan Metabolite Analysis Engineering Technology Center of Cells for Medical Use, Guangdong Xinghai Institute of Cell, Dongguan, China
- Key Laboratory of Medical Bioactive Molecular Research for Department of Education of Guangdong Province, Collaborative Innovation Center for Antitumor Active Substance Research and Development, Zhanjiang, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
27
|
Vasquez R, Oh JK, Song JH, Kang DK. Gut microbiome-produced metabolites in pigs: a review on their biological functions and the influence of probiotics. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2022; 64:671-695. [PMID: 35969697 PMCID: PMC9353353 DOI: 10.5187/jast.2022.e58] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/23/2022] [Accepted: 07/04/2022] [Indexed: 11/20/2022]
Abstract
The gastrointestinal tract is a complex ecosystem that contains a large number of microorganisms with different metabolic capacities. Modulation of the gut microbiome can improve the growth and promote health in pigs. Crosstalk between the host, diet, and the gut microbiome can influence the health of the host, potentially through the production of several metabolites with various functions. Short-chain and branched-chain fatty acids, secondary bile acids, polyamines, indoles, and phenolic compounds are metabolites produced by the gut microbiome. The gut microbiome can also produce neurotransmitters (such as γ-aminobutyric acid, catecholamines, and serotonin), their precursors, and vitamins. Several studies in pigs have demonstrated the importance of the gut microbiome and its metabolites in improving growth performance and feed efficiency, alleviating stress, and providing protection from pathogens. The use of probiotics is one of the strategies employed to target the gut microbiome of pigs. Promising results have been published on the use of probiotics in optimizing pig production. This review focuses on the role of gut microbiome-derived metabolites in the performance of pigs and the effects of probiotics on altering the levels of these metabolites.
Collapse
Affiliation(s)
- Robie Vasquez
- Department of Animal Resources Science,
Dankook University, Cheonan 31116, Korea
| | - Ju Kyoung Oh
- Department of Animal Resources Science,
Dankook University, Cheonan 31116, Korea
| | - Ji Hoon Song
- Department of Animal Resources Science,
Dankook University, Cheonan 31116, Korea
| | - Dae-Kyung Kang
- Department of Animal Resources Science,
Dankook University, Cheonan 31116, Korea
| |
Collapse
|
28
|
Liu G, Zheng J, Gu K, Wu C, Jia G, Zhao H, Chen X, Wang J. Calcium-sensing receptor protects intestinal integrity and alleviates the inflammatory response via the Rac1/PLCγ1 signaling pathway. Anim Biotechnol 2021:1-14. [PMID: 34762003 DOI: 10.1080/10495398.2021.1998090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
This study aimed to test the hypothesis that the calcium-sensing receptor (CaSR) can protect intestinal epithelial barrier integrity and decrease inflammatory response mediated by the Ras-related C3 botulinum toxin substrate 1 (Rac1)/phospholipase Cγ1 (PLC-γ1) signaling pathway. IPEC-J2 monolayers were treated without or with TNF-α in the absence or presence of CaSR antagonist (NPS 2143), CaSR overexpression, and Rac1 silencing, PLCγ1 silencing or spermine. Results showed that spermine increased transepithelial electrical resistance (TER), tight junction protein levels, the protein concentration of Rac1/PLC-γ1 signaling pathway, and decreased paracellular permeability in the presence of TNF-α. NPS2143 inhibited spermine-induced change in above-mentioned parameters. CaSR overexpression increased TER, the levels of tight junction proteins and the protein concentration of CaSR, phosphorylated PLCγ1, Rac1, and IP3, and decreased paracellular permeability and contents of interleukin-8 (IL-8) and TNF-α after TNF-α challenge. Rac1 and PLCγ1 silencing inhibited CaSR-induced increase in barrier function and the protein concentration of phosphorylated PLCγ1, Rac1, and IP3, and decrease in contents of IL-8 and TNF-α after TNF-α challenge. These results suggest that CaSR activation protects intestinal integrity and alleviates the inflammatory response by activating Rac1 and PLCγ1 signaling after TNF-α challenge, and spermine can maintain barrier function via CaSR/Rac1/PLC-γ1 pathway.
Collapse
Affiliation(s)
- Guangmang Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, Sichuan, China.,Key laboratory of Animal Disease-resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China
| | - Jie Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, Sichuan, China.,Key laboratory of Animal Disease-resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China
| | - Ke Gu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, Sichuan, China.,Key laboratory of Animal Disease-resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China
| | - Caimei Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, Sichuan, China.,Key laboratory of Animal Disease-resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China
| | - Gang Jia
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, Sichuan, China.,Key laboratory of Animal Disease-resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China
| | - Hua Zhao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, Sichuan, China.,Key laboratory of Animal Disease-resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China
| | - Xiaoling Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, Sichuan, China.,Key laboratory of Animal Disease-resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China
| | - Jing Wang
- Maize Research Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
29
|
Cookson TA. Bacterial-Induced Blood Pressure Reduction: Mechanisms for the Treatment of Hypertension via the Gut. Front Cardiovasc Med 2021; 8:721393. [PMID: 34485420 PMCID: PMC8414577 DOI: 10.3389/fcvm.2021.721393] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 07/16/2021] [Indexed: 01/08/2023] Open
Abstract
Hypertension is a major risk factor for the development of cardiovascular disease. As more research into the gut microbiome emerges, we are finding increasing evidence to support that these microbes may have significant positive and negative effects on blood pressure and associated disorders. The bacterial-derived metabolites that are produced in the gut are capable of widespread effects to several tissue types and organs in the body. It is clear that the extensive metabolic function that is lost with gut dysbiosis is unlikely to be replenished with a single metabolite or bacterial strain. Instead, combinations of bacteria and concomitant therapies will provide a more well-rounded solution to manage hypertension. The bioactive molecules that are recognized in this review will inform on ideal characteristics of candidate bacteria and provide direction for future research on the gut microbiome in hypertension.
Collapse
|
30
|
Zheng J, Liu G, Wu C, Jia G, Zhao H, Chen X, Wang J. Effect of calcium-sensing receptor on the migration and proliferation of porcine intestinal epithelial cells. Anim Biotechnol 2021; 34:365-374. [PMID: 34459707 DOI: 10.1080/10495398.2021.1968885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The rapid healing of impaired intestinal surface plays a role in maintaining intestinal homeostasis. This study investigated the effect of calcium-sensing receptor (CaSR) on the migration and proliferation of intestinal porcine epithelial cells (IPEC-J2). Results showed that cell migration area and width were increased by R568 (CaSR activator) and decreased by NPS2143 (CaSR inhibitor). The protein level of GTP-rac1 and the phosphorylation of phospholipase C gamma 1 (PLCγ1) were increased by 2 µM R568. Furthermore, R568 + 120 µM NSC23766 (Rac1 inhibitor) and R568 + 1 µM U73122 (PLCγ1 inhibitor) decreased the protein level of GTP-rac1 and the phosphorylated PLCγ1, respectively, and both inhibited cell migration compared with R568. In addition, spermine increased the protein expression levels of CaSR and the levels of GTP-rac1 and the phosphorylated PLCγ1 and thereby promoted the migration of IPEC-J2 cells. Moreover, R568 improved the proliferation of the IPEC-J2 cells. Spermine increased cell proliferation, but NPS2143 incubated with spermine decreased cell proliferation compared with the spermine group. This study suggests that CaSR activation increased cell migration by activating Rac1 and PLCγ1 signaling and improved cell proliferation, and both effects were regulated by spermine by activating CaSR.
Collapse
Affiliation(s)
- Jie Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, Sichuan, China.,Key laboratory of Animal Disease-resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China
| | - Guangmang Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, Sichuan, China.,Key laboratory of Animal Disease-resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China
| | - Caimei Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, Sichuan, China.,Key laboratory of Animal Disease-resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China
| | - Gang Jia
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, Sichuan, China.,Key laboratory of Animal Disease-resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China
| | - Hua Zhao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, Sichuan, China.,Key laboratory of Animal Disease-resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China
| | - Xiaoling Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, Sichuan, China.,Key laboratory of Animal Disease-resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, Sichuan, China
| | - Jing Wang
- Maize Research Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
31
|
Xu X, Liu G, Jia G, Zhao H, Chen X, Wu C, Wang J. Effects of spermine on the proliferation and migration of porcine intestinal epithelial cells. Anim Biotechnol 2021; 34:253-260. [PMID: 34369303 DOI: 10.1080/10495398.2021.1955699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Whether spermine promotes the repair of porcine intestinal epithelium damage through Ras-related C3 botulinum toxin substrate 1 (Rac1)/phospholipase C-γ1 (PLC-γ1) signaling remains unclear. The current study investigated the effects of spermine addition on the proliferation and migration of IPEC-J2 cells and the effects of Rac1/PLC-γ1 signaling on cell migration. We showed that the inhibitors of Rac1 (NSC-23766) and PLC-γ1 (U73122) reduced cell migration and decreased the protein levels of Rac1 and PLC-γ1 in the cells. Moreover, spermine promoted the proliferation and migration of the IPEC-J2 cells, that is, 1 µM spermine exhibited the best effect, and spermine treatment increased the protein levels of Rac1 and PLC-γ1. Further experiments showed that spermine treatment increased cell migration and enhanced Rac1 and PLC-γ1 protein levels, compared with NSC-23766 and U73122 treatments with spermine. In conclusion, spermine treatment promoted the repair of damaged porcine intestinal epithelium by accelerating cell proliferation and migration mediated by Rac1/PLC-γ1 signaling.
Collapse
Affiliation(s)
- Xiaomei Xu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China.,Key laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Guangmang Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China.,Key laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Gang Jia
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China.,Key laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Hua Zhao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China.,Key laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Xiaoling Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China.,Key laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Caimei Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China.,Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China.,Key laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Jing Wang
- Maize Research Institute, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
32
|
Cai L, Wei Z, Zhao X, Li Y, Li X, Jiang X. Gallic acid mitigates LPS-induced inflammatory response via suppressing NF-κB signalling pathway in IPEC-J2 cells. J Anim Physiol Anim Nutr (Berl) 2021; 106:1000-1008. [PMID: 34288130 DOI: 10.1111/jpn.13612] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 05/12/2021] [Accepted: 07/04/2021] [Indexed: 01/26/2023]
Abstract
Gallic acid is a phenolic compound that exhibits antibacterial, antioxidative and anti-inflammatory functions. In a previous study, we found that dietary supplementation with gallic acid decreased incidence of diarrhoea and protected intestinal integrity in weaning piglets. However, the underlying mechanism remains unclear. Here, a pig intestinal epithelial cell line (IPEC-J2) was used as an in vitro model to explore the antioxidant and anti-inflammatory capacity of gallic acid. IPEC-J2 cells were stimulated with hydrogen peroxide (H2 O2 ) and lipopolysaccharide (LPS) to establish oxidative and inflammatory models, respectively. Results showed that H2 O2 significantly decreased catalase (CAT) secretion and CAT mRNA abundance in the cells (p < 0.05), while pretreatment with gallic acid did not prevent the decrease in CAT expression induced by H2 O2 . However, gallic acid pretreatment mitigated the increased expression of the tumour necrosis factor-α and interleukin-8 genes caused by LPS in IPEC-J2 cells (p < 0.05). In addition, pretreatment with gallic acid significantly suppressed phosphorylation of NF-κB and IκBα in LPS-stimulated IPEC-J2 cells. Moreover, LPS stimulation decreased the protein abundance of zona occludens 1 (ZO-1) and occludin, while pretreatment with gallic acid preserved expression level of tight junction proteins ZO-1 and occludin in LPS-stimulated IPEC-J2 cells (p < 0.05). In conclusion, gallic acid may mitigate LPS-induced inflammatory responses by inhibiting the NF-κB signalling pathway, exerting positive effects on the barrier function of IPEC-J2 cells.
Collapse
Affiliation(s)
- Long Cai
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zixi Wei
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xuemei Zhao
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yanpin Li
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xilong Li
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xianren Jiang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
33
|
Wang H, Xia P, Lu Z, Su Y, Zhu W. Metabolome-Microbiome Responses of Growing Pigs Induced by Time-Restricted Feeding. Front Vet Sci 2021; 8:681202. [PMID: 34239912 PMCID: PMC8258120 DOI: 10.3389/fvets.2021.681202] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 05/20/2021] [Indexed: 01/25/2023] Open
Abstract
Time-restricted feeding (TRF) mode is a potential strategy in improving the health and production of farm animals. However, the effect of TRF on microbiota and their metabolism in the large intestine of the host remains unclear. Therefore, the present study aimed to investigate the responses of microbiome and metabolome induced by TRF based on a growing-pig model. Twelve crossbred growing barrows were randomly allotted into two groups with six replicates (1 pig/pen), namely, the free-access feeding group (FA) and TRF group. Pigs in the FA group were fed free access while the TRF group were fed free access within a regular time three times per day at 07:00–08:00, 12:00–13:00, and 18:00–19:00, respectively. Results showed that the concentrations of NH4-N, putrescine, cadaverine, spermidine, spermine, total biogenic amines, isobutyrate, butyrate, isovalerate, total SCFA, and lactate were increased while the pH value in the colonic digesta and the concentration of acetate was decreased in the TRF group. The Shannon index was significantly increased in the TRF group; however, no significant effects were found in the Fisher index, Simpson index, ACE index, Chao1 index, and observed species between the two groups. In the TRF group, the relative abundances of Prevotella 1 and Eubacterium ruminantium group were significantly increased while the relative abundances of Clostridium sensu sticto 1, Lactobacillus, and Eubacterium coprostanoligenes group were decreased compared with the FA group. PLS-DA analysis revealed an obvious and regular variation between the FA and TRF groups, further pathway enrichment analysis showed that these differential features were mainly enriched in pyrimidine metabolism, nicotinate and nicotinamide metabolism, glycerolipid metabolism, and fructose and mannose metabolism. In addition, Pearson's correlation analysis indicated that the changes in the microbial genera were correlated with the colonic metabolites. In conclusion, these results together indicated that although the overall microbial composition in the colon was not changed, TRF induced the gradient changes of the nutrients and metabolites which were correlated with certain microbial genera including Lactobacillus, Eubacterium_ruminantium group, Eubacterium coprostanoligenes group, Prevotella 1, and Clostridium sensu sticto 1. However, more studies are needed to understand the impacts of TRF on the health and metabolism of growing pigs.
Collapse
Affiliation(s)
- Hongyu Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Pengke Xia
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Zhiyang Lu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Yong Su
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
34
|
Huang C, Fan Z, Han D, Johnston LJ, Ma X, Wang F. Pyrroloquinoline quinone regulates the redox status in vitro and in vivo of weaned pigs via the Nrf2/HO-1 pathway. J Anim Sci Biotechnol 2021; 12:77. [PMID: 34140030 PMCID: PMC8212497 DOI: 10.1186/s40104-021-00595-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/11/2021] [Indexed: 12/28/2022] Open
Abstract
Background Oxidative stress is a main cause of piglet gut damage and diarrhea. Pyrroloquinoline quinone (PQQ), is a novel redox cofactor with antioxidant properties. However, the effect and mechanism that PQQ supplementation decreases oxidative injury in weaned pigs is not understood. Therefore, the aim of this study is to confirm the effect of PQQ on regulating redox status in weaned pigs and the mechanism for antioxidant function by porcine intestinal epithelial cell line (IPEC-J2) challenged with H2O2. Results Experiment 1, 144 Duroc × Landrace × Yorkshire pigs (weaned at 28 d) were allocated to four groups: received a basal diet (control) and diets supplemented with 0.15%, 0.30% and 0.45% PQQ, respectively. On d 28, growth performance, diarrhea incidence and redox factors were measured. Experiment 2, IPEC-J2 were treated with or without PQQ in the presence or absence of H2O2 for indicated time points. Experiment 3, IPEC-J2 were transfected with or without Nrf2 siRNA, then treated according to Experiment 2. The cell viability, redox factors, protein of tight junctions and Nrf2 pathway were determined. In vivo, PQQ supplementation demonstrated dose-related improvements in average daily gain, and gain to feed ratio (Linear P < 0.05). During d 0–28, compared to controls, 0.45% PQQ supplementation for pigs decreased diarrhea incidence and MDA content in liver and jejunum, and increased concentration of SOD in liver; 0.3% PQQ supplementation decreased ileal and liver MDA concentration; and 0.15% PQQ supplementation decreased ileal MDA concentration (P < 0.05). In vitro, compared to cells cultured with H2O2, pre-treatment with PQQ increased cell viability, tight junction proteins expression including ZO-1, ZO-2, Occludin and Claudin-1; and decreased ROS concentration and level of Caspase-3 (P < 0.05); as well as upregulated the ratio of Bcl-2 to Bax and protein expression of nuclear Nrf2, HO-1. Notably, Nrf2 knockdown by transfection with Nrf2 siRNA largely abrogated the positive effects of PQQ pretreatment on H2O2-induced intracellular changes. Conclusions PQQ administration attenuated oxidative stress in weaned pigs which is associated with activation of Nrf2/HO-1 pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s40104-021-00595-x.
Collapse
Affiliation(s)
- Caiyun Huang
- State Key Lab of Animal Nutrition, College of Animal Science & Technology, China Agricultural University, Beijing, 100193, China
| | - Zijuan Fan
- State Key Lab of Animal Nutrition, College of Animal Science & Technology, China Agricultural University, Beijing, 100193, China
| | - Dandan Han
- State Key Lab of Animal Nutrition, College of Animal Science & Technology, China Agricultural University, Beijing, 100193, China
| | - Lee J Johnston
- Swine Nutrition and Production, West Central Research and Outreach Center, University of Minnesota, Morris, MN, USA
| | - Xi Ma
- State Key Lab of Animal Nutrition, College of Animal Science & Technology, China Agricultural University, Beijing, 100193, China.,Department of Internal Medicine/Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Fenglai Wang
- State Key Lab of Animal Nutrition, College of Animal Science & Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
35
|
Yurdagul A, Kong N, Gerlach BD, Wang X, Ampomah P, Kuriakose G, Tao W, Shi J, Tabas I. ODC (Ornithine Decarboxylase)-Dependent Putrescine Synthesis Maintains MerTK (MER Tyrosine-Protein Kinase) Expression to Drive Resolution. Arterioscler Thromb Vasc Biol 2021; 41:e144-e159. [PMID: 33406854 PMCID: PMC8034502 DOI: 10.1161/atvbaha.120.315622] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/22/2020] [Indexed: 11/16/2022]
Abstract
OBJECTIVE ODC (ornithine decarboxylase)-dependent putrescine synthesis promotes the successive clearance of apoptotic cells (ACs) by macrophages, contributing to inflammation resolution. However, it remains unknown whether ODC is required for other arms of the resolution program. Approach and Results: RNA sequencing of ODC-deficient macrophages exposed to ACs showed increases in mRNAs associated with heightened inflammation and decreases in mRNAs related to resolution and repair compared with WT (wild type) macrophages. In zymosan peritonitis, myeloid ODC deletion led to delayed clearance of neutrophils and a decrease in the proresolving cytokine, IL (interleukin)-10. Nanoparticle-mediated silencing of macrophage ODC in a model of atherosclerosis regression lowered IL-10 expression, decreased efferocytosis, enhanced necrotic core area, and reduced fibrous cap thickness. Mechanistically, ODC deletion lowered basal expression of MerTK (MER tyrosine-protein kinase)-an AC receptor-via a histone methylation-dependent transcriptional mechanism. Owing to lower basal MerTK, subsequent exposure to ACs resulted in lower MerTK-Erk (extracellular signal-regulated kinase) 1/2-dependent IL-10 production. Putrescine treatment of ODC-deficient macrophages restored the expression of both MerTK and AC-induced IL-10. CONCLUSIONS These findings demonstrate that ODC-dependent putrescine synthesis in macrophages maintains a basal level of MerTK expression needed to optimally resolve inflammation upon subsequent AC exposure. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Arif Yurdagul
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Brennan D. Gerlach
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Patrick Ampomah
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - George Kuriakose
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
- Department of Physiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
36
|
Yang KL, Lejeune A, Chang G, Scher JU, Koralov SB. Microbial-derived antigens and metabolites in spondyloarthritis. Semin Immunopathol 2021; 43:163-172. [PMID: 33569635 DOI: 10.1007/s00281-021-00844-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/20/2021] [Indexed: 12/30/2022]
Abstract
Spondyloarthritis (SpA) is a group of chronic, immune-mediated, inflammatory diseases affecting the bone, synovium, and enthesis. Microbiome, the community of microorganisms that has co-evolved with human hosts, plays a pivotal role in human health and disease. This invisible "essential organ" supplies the host with a myriad of chemicals and molecules. In turn, microbial metabolites can serve as messengers for microbes to communicate with each other and in the cross-talk with host cells. Gut dysbiosis in SpA is associated with altered microbial metabolites, and an accumulated body of research has contributed to the understanding that changes in intestinal microbiota can modulate disease pathogenesis. We review the novel findings from human and animal studies to provide an overview of the contribution of individual microbial metabolites and antigens to SpA.
Collapse
Affiliation(s)
- Katharine Lu Yang
- Department of Pathology, NYU School of Medicine, 522 First Ave. Smilow Research Bldg 511, New York, NY, 10016, USA
| | - Alannah Lejeune
- Department of Pathology, NYU School of Medicine, 522 First Ave. Smilow Research Bldg 511, New York, NY, 10016, USA
| | - Gregory Chang
- Department of Radiology, NYU School of Medicine, New York, NY, 10016, USA
| | - Jose U Scher
- Division of Rheumatology, Department of Medicine, NYU School of Medicine, New York, NY, 10016, USA. .,Division of Rheumatology and Psoriatic Arthritis Center, 301 East 17th St, Room 1608, New York, NY, 10003, USA.
| | - Sergei B Koralov
- Department of Pathology, NYU School of Medicine, 522 First Ave. Smilow Research Bldg 511, New York, NY, 10016, USA.
| |
Collapse
|
37
|
Ren W, Bin P, Yin Y, Wu G. Impacts of Amino Acids on the Intestinal Defensive System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1265:133-151. [PMID: 32761574 DOI: 10.1007/978-3-030-45328-2_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The intestine interacts with a diverse community of antigens and bacteria. To keep its homeostasis, the gut has evolved with a complex defense system, including intestinal microbiota, epithelial layer and lamina propria. Various factors (e.g., nutrients) affect the intestinal defensive system and progression of intestinal diseases. This review highlights the current understanding about the role of amino acids (AAs) in protecting the intestine from harm. Amino acids (e.g., arginine, glutamine and tryptophan) are essential for the function of intestinal microbiota, epithelial cells, tight junction, goblet cells, Paneth cells and immune cells (e.g., macrophages, B cells and T cells). Through the modulation of the intestinal defensive system, AAs maintain the integrity and function of the intestinal mucosa and inhibit the progression of various intestinal diseases (e.g., intestinal infection and intestinal colitis). Thus, adequate intake of functional AAs is crucial for intestinal and whole-body health in humans and other animals.
Collapse
Affiliation(s)
- Wenkai Ren
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Peng Bin
- Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product, Safety of Ministry of Education of China, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yulong Yin
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, USA.
| |
Collapse
|