1
|
Rose EC, Simon JM, Gomez-Martinez I, Magness ST, Odle J, Blikslager AT, Ziegler AL. Single-cell transcriptomics predict novel potential regulators of acute epithelial restitution in the ischemia-injured intestine. Am J Physiol Gastrointest Liver Physiol 2025; 328:G182-G196. [PMID: 39853303 DOI: 10.1152/ajpgi.00194.2024] [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: 07/02/2024] [Revised: 08/05/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025]
Abstract
Intestinal ischemic injury damages the epithelial barrier and predisposes patients to life-threatening sepsis unless that barrier is rapidly restored. There is an age dependency in intestinal recovery in that neonates are the most susceptible to succumb to disease of the intestinal barrier compared with older patients. We have developed a pig model that demonstrates age-dependent failure of intestinal barrier restitution in neonatal pigs, which can be rescued by the direct application of juvenile pig mucosal tissue, but the mechanisms of rescue remain undefined. We hypothesized that by identifying a subpopulation of restituting enterocytes by their expression of cell migration transcriptional pathways, we can then predict novel upstream regulators of age-dependent restitution response programs. Superficial mucosal epithelial cells from recovering ischemic jejunum of juvenile pigs underwent single-cell transcriptomics and the predicted upstream regulator, colony stimulating factor-1 (CSF-1), was interrogated in our model. A subcluster of absorptive enterocytes expressed several cell migration pathways key to restitution. Differentially expressed genes in this subcluster predicted their upstream regulation by colony stimulating factor-1 (CSF-1). We validated age-dependent induction of CSF-1 by ischemia and documented that CSF-1 and colony-stimulating factor-1 receptor (CSF1R) co-localized in ischemic juvenile, but not neonatal, wound-adjacent epithelial cells and in the restituted epithelium of juveniles and rescued neonates. Furthermore, the CSF-1 blockade reduced restitution in vitro, and CSF-1 improved barrier function in injured neonatal pigs in preliminary ex vivo experiments. These studies validate an approach to inform potential novel therapeutic targets, such as CSF-1, to improve outcomes in neonates with intestinal injury in a unique pig model.NEW & NOTEWORTHY These studies validate an approach to identify and predict upstream regulation of restituting epithelium in a unique pig intestinal ischemic injury model. Identification of potential molecular mediators of restitution, such as CSF-1, will inform the development of targeted therapeutic interventions for the medical management of patients with ischemia-mediated intestinal injury.
Collapse
Affiliation(s)
- Elizabeth C Rose
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Jeremy M Simon
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Ismael Gomez-Martinez
- Bioinformatics and Analytics Research Collaborative, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Scott T Magness
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Jack Odle
- Department of Animal Science, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, North Carolina, United States
| | - Anthony T Blikslager
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Amanda L Ziegler
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| |
Collapse
|
2
|
Schokker D, van den Esker MH, Bossers A, Allaart JG, van Hees H, de Greeff A, Rebel JMJ. Neonatal and maternal dietary interventions driving microbiota and functionality in piglet gut compartments. Sci Rep 2025; 15:6771. [PMID: 40000703 PMCID: PMC11861315 DOI: 10.1038/s41598-025-90781-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Feed additives aiming to improve gastrointestinal health are frequently supplied to piglets after weaning (d28) but might be more effective when administered before weaning. In this period, feed additives can either be administered directly to neonates, or indirectly via sow's feed. It is yet unknown what the effect of the administration route is on gut functionality and health in the piglets. Therefore, we compared the effect of different dietary interventions on gut functionality after maternal administration (lactation feed) to the neonatal administration route (oral gavage). These feed interventions included medium chain fatty acids (MCFA), beta-glucans (BG), and galacto-oligosaccharides (GOS). For the maternal administration route, MCFA showed a significant difference in alpha diversity parameter, observed species at d1 and one differentially expressed gene (DEG), and 99 DEG at d31. Pathway enrichment analysis showed association to immune processes and metabolism. For BG, only 21 DEG were observed at d31, these DEGs were associated to signal transduction and sympathetic nerve pathway. For GOS, 816 DEG were observed for GOS at d1, and 77 at d31, where DEGs at d1 were associated to immune processes. For the neonatal administration route, MCFA showed 94 DEG and GOS 6 DEG. Where DEGs in MCFA were mainly associated to cell adhesion processes. When comparing the administration routes directly between treatment groups, we observed significant differences in alpha diversity parameters, observed species at d31 for MCFA, Shannon for GOS, as well as for beta diversity in GOS. For MCFA 515 DEG were observed, for BG 503 DEG, and for GOS 996 DEG. Where for MCFA most pathways were associated to immunological processes, BG showed more metabolism, and GOS mainly metabolism with a few immunological processes. The type of intervention and the administration route influence gut functionality of the piglets. MCFA administration led to a more differentially orchestrated response when comparing the neonatal and maternal administration route then the other two additives. This implies that for each nutritional intervention in early life of a pig the optimal route of administration needs to be determined.
Collapse
Affiliation(s)
- Dirkjan Schokker
- Wageningen Bioveterinary Research, Lelystad, The Netherlands.
- , Postbus 338, Wageningen, 6700 AH, The Netherlands.
| | | | - Alex Bossers
- Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | | | - Hubèrt van Hees
- Trouw Nutrition Research and Development, Boxmeer, The Netherlands
| | | | | |
Collapse
|
3
|
Archontakis-Barakakis P, Mavridis T, Chlorogiannis DD, Barakakis G, Laou E, Sessler DI, Gkiokas G, Chalkias A. Intestinal oxygen utilisation and cellular adaptation during intestinal ischaemia-reperfusion injury. Clin Transl Med 2025; 15:e70136. [PMID: 39724463 DOI: 10.1002/ctm2.70136] [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: 08/28/2024] [Revised: 11/06/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
The gastrointestinal tract can be deranged by ailments including sepsis, trauma and haemorrhage. Ischaemic injury provokes a common constellation of microscopic and macroscopic changes that, together with the paradoxical exacerbation of cellular dysfunction and death following restoration of blood flow, are collectively known as ischaemia-reperfusion injury (IRI). Although much of the gastrointestinal tract is normally hypoxemic, intestinal IRI results when there is inadequate oxygen availability due to poor supply (pathological hypoxia) or abnormal tissue oxygen use and metabolism (dysoxia). Intestinal oxygen uptake usually remains constant over a wide range of blood flows and pressures, with cellular function being substantively compromised when ischaemia leads to a >50% decline in intestinal oxygen consumption. Restoration of perfusion and oxygenation provokes additional injury, resulting in mucosal damage and disruption of intestinal barrier function. The primary cellular mechanism for sensing hypoxia and for activating a cascade of cellular responses to mitigate the injury is a family of heterodimer proteins called hypoxia-inducible factors (HIFs). The HIF system is connected to numerous biochemical and immunologic pathways induced by IRI and the concentration of those proteins increases during hypoxia and dysoxia. Activation of the HIF system leads to augmented transcription of specific genes in various types of affected cells, but may also augment apoptotic and inflammatory processes, thus aggravating gut injury. KEY POINTS: During intestinal ischaemia, mitochondrial oxygen uptake is reduced when cellular oxygen partial pressure decreases to below the threshold required to maintain normal oxidative metabolism. Upon reperfusion, intestinal hypoxia may persist because microcirculatory flow remains impaired and/or because available oxygen is consumed by enzymes, intestinal cells and neutrophils.
Collapse
Affiliation(s)
| | - Theodoros Mavridis
- Department of Neurology, Tallaght University Hospital (TUH)/The Adelaide and Meath Hospital incorporating the National Children's Hospital (AMNCH), Dublin, Ireland
| | | | - Georgios Barakakis
- Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleni Laou
- Department of Anesthesiology, Agia Sophia Children's Hospital, Athens, Greece
| | - Daniel I Sessler
- Center for Outcomes Research and Department of Anesthesiology, UTHealth, Houston, Texas, USA
- Outcomes Research Consortium®, Houston, Texas, USA
| | - George Gkiokas
- Second Department of Surgery, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios Chalkias
- Outcomes Research Consortium®, Houston, Texas, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Critical Care Medicine, Tzaneio General Hospital, Piraeus, Greece
| |
Collapse
|
4
|
Rose EC, Simon JM, Gomez-Martinez I, Magness ST, Odle J, Blikslager AT, Ziegler AL. Single-cell transcriptomics predict novel potential regulators of acute epithelial restitution in the ischemia-injured intestine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601271. [PMID: 38979337 PMCID: PMC11230382 DOI: 10.1101/2024.06.28.601271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Intestinal ischemic injury damages the epithelial barrier predisposes patients to life-threatening sepsis unless that barrier is rapidly restored. There is an age-dependency of intestinal recovery in that neonates are the most susceptible to succumb to disease of the intestinal barrier versus older patients. We have developed a pig model that demonstrates age-dependent failure of intestinal barrier restitution in neonatal pigs which can be rescued by the direct application of juvenile pig mucosal tissue, but the mechanisms of rescue remain undefined. We hypothesized that by identifying a subpopulation of restituting enterocytes by their expression of cell migration transcriptional pathways, we can then predict novel upstream regulators of age-dependent restitution response programs. Superficial mucosal epithelial cells from recovering ischemic jejunum of juvenile pigs were processed for single cell RNA sequencing analysis, and predicted upstream regulators were assessed in a porcine intestinal epithelial cell line (IPEC-J2) and banked tissues. A subcluster of absorptive enterocytes expressed several cell migration pathways key to restitution. Differentially expressed genes in this subcluster predicted their upstream regulation included colony stimulating factor-1 (CSF-1). We validated age-dependent induction of CSF-1 by ischemia and documented that CSF-1 and CSF1R co-localized in ischemic juvenile, but not neonatal, wound-adjacent epithelial cells and in the restituted epithelium of juveniles and rescued (but not control) neonates. Further, the CSF1R inhibitor BLZ945 reduced restitution in scratch wounded IPEC-J2 cells. These studies validate an approach to inform potential novel therapeutic targets, such as CSF-1, to improve outcomes in neonates with intestinal injury in a unique pig model. NEW & NOTEWORTHY These studies validate an approach to identify and predict upstream regulation of restituting epithelium in a unique pig intestinal ischemic injury model. Identification of potential molecular mediators of restitution, such as CSF-1, will inform the development of targeted therapeutic interventions for medical management of patients with ischemia-mediated intestinal injury.
Collapse
|
5
|
Baker JT, Duarte ME, Kim SW. Effects of dietary xylanase supplementation on growth performance, intestinal health, and immune response of nursery pigs fed diets with reduced metabolizable energy. J Anim Sci 2024; 102:skae026. [PMID: 38280204 PMCID: PMC10889732 DOI: 10.1093/jas/skae026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/25/2024] [Indexed: 01/29/2024] Open
Abstract
This study aimed to investigate the effects of xylanase on growth performance and intestinal health of nursery pigs fed diets with reduced metabolizable energy (ME). One hundred ninety-two pigs at 8.7 kg ± 0.7 body weight (BW) after 7 d of weaning were allotted in a randomized complete block design with initial BW and sex as blocks. Eight dietary treatments consisted of 5 ME levels (3,400, 3,375, 3,350, 3,325, and 3,300 kcal ME/kg) below the NRC (2012) requirement and 4 levels of xylanase (0, 1,200, 2,400, and 3,600 XU/kg) to a diet with 3,300 kcal ME/kg. All pigs received their respective treatments for 35 d in 2 phases, pre-starter (14 d) and starter (21 d). On day 35, eight pigs in 3,400 kcal/kg (CON), 3,300 kcal/kg (LE), and 3,300 kcal/kg + 3,600 XU xylanase/kg (LEX) were euthanized to collect jejunal tissues and digesta for the evaluation of mucosa-associated microbiota, intestinal immune response, oxidative stress status, intestinal morphology, crypt cell proliferation, and digesta viscosity as well as ileal digesta to measure apparent ileal digestibility. Data were analyzed using the MIXED procedure on SAS 9.4. The LE increased (P < 0.05) jejunal digesta viscosity, tended to have decreased (P = 0.053) relative abundance of Prevotella, and tended to increase (P = 0.055) Lactobacillus. The LE also increased (P < 0.05) the concentration of protein carbonyl whereas malondialdehyde, villus height (VH), villus height to crypt depth ratio (VH:CD), apparent ileal digestibility (AID) of nutrients, and finally average daily feed intake were decreased (P < 0.05). The LE did not affect average daily gain (ADG). The LEX decreased (P < 0.05) digesta viscosity, increased (P < 0.05) the relative abundance of Prevotella, decreased (P < 0.05) Helicobacter, decreased (P < 0.05) the concentration of protein carbonyl, tended to increase (P = 0.065) VH, and decreased (P < 0.05) VH:CD and crypt cell proliferation. Moreover, LEX increased (P < 0.05) the AID of dry matter and gross energy and tended to increase (P = 0.099; P = 0.076) AID of crude protein, and ether extract. The LEX did not affect ADG but did tend to decrease (P = 0.070) fecal score during the starter phase. Overall, reducing ME negatively affected intestinal health parameters and nutrient digestibility without affecting growth. Supplementation of xylanase mitigated some of the negative effects observed by ME reduction on intestinal health and digestibility of nutrients without affecting growth.
Collapse
Affiliation(s)
- Jonathan T Baker
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Marcos Elias Duarte
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
6
|
Ma T, Huang W, Li Y, Jin H, Kwok LY, Sun Z, Zhang H. Probiotics alleviate constipation and inflammation in late gestating and lactating sows. NPJ Biofilms Microbiomes 2023; 9:70. [PMID: 37741814 PMCID: PMC10517943 DOI: 10.1038/s41522-023-00434-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/11/2023] [Indexed: 09/25/2023] Open
Abstract
Constipation and systemic inflammation are common in late pregnant and lactating sows, which cause health problems like uteritis, mastitis, dystocia, or even stillbirth, further influencing piglets' survival and growth. Probiotic supplementation can improve such issues, but the beneficial mechanism of relieving constipation and enhancing gut motility remains underexplored. This study aimed to investigate the effects and mechanism of probiotic supplementation in drinking water to late pregnant sows on constipation, inflammation, and piglets' growth performance. Seventy-four sows were randomly allocated to probiotic (n = 36) and control (n = 38) groups. Probiotic treatment significantly relieved sow constipation, enhanced serum IL-4 and IL-10 levels while reducing serum IL-1β, IL-12p40, and TNF-α levels, and increased piglet daily gain and weaning weight. Furthermore, probiotic administration reshaped the sow gut bacteriome and phageome structure/diversity, accompanied by increases in some potentially beneficial bacteria. At 113 days of gestation, the probiotic group was enriched in several gut microbial bioactive metabolites, multiple carbohydrate-active enzymes that degrade pectin and starch, fecal butyrate and acetate, and some serum metabolites involved in vitamin and amino acid metabolism. Our integrated correlation network analysis revealed that the alleviation of constipation and inflammation was associated with changes in the sow gut bacteriome, phageome, bioactive metabolic potential, and metabolism.
Collapse
Affiliation(s)
- Teng Ma
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Weiqiang Huang
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Yalin Li
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Hao Jin
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Lai-Yu Kwok
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Zhihong Sun
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Heping Zhang
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China.
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China.
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China.
| |
Collapse
|
7
|
Eudy BJ, Odle J, Lin X, Maltecca C, Walter KR, McNulty NP, Fellner V, Jacobi SK. Dietary Prebiotic Oligosaccharides and Arachidonate Alter the Fecal Microbiota and Mucosal Lipid Composition of Suckling Pigs. J Nutr 2023; 153:2249-2262. [PMID: 37348760 DOI: 10.1016/j.tjnut.2023.06.019] [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: 04/13/2023] [Revised: 06/09/2023] [Accepted: 06/15/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Early intestinal development is important to infant vitality, and optimal formula composition can promote gut health. OBJECTIVES The objectives were to evaluate the effects of arachidonate (ARA) and/or prebiotic oligosaccharide (PRE) supplementation in formula on the development of the microbial ecosystem and colonic health parameters. METHODS Newborn piglets were fed 4 formulas containing ARA [0.5 compared with 2.5% of dietary fatty acids (FAs)] and PRE (0 compared with 8 g/L, containing a 1:1 mixture of galactooligosaccharides and polydextrose) in a 2 x 2 factorial design for 22 d. Fecal samples were collected weekly and analyzed for relative microbial abundance. Intestinal samples were collected on day 22 and analyzed for mucosal FAs, pH, and short-chain FAs (SCFAs). RESULTS PRE supplementation significantly increased genera within Bacteroidetes and Firmicutes, including Anaerostipes, Mitsuokella, Prevotella, Clostridium IV, and Bulleidia, and resulted in progressive separation from controls as determined by Principal Coordinates Analysis. Concentrations of SCFA increased from 70.98 to 87.37 mM, with an accompanying reduction in colonic pH. ARA supplementation increased the ARA content of the colonic mucosa from 2.35-5.34% of total FAs. PRE supplementation also altered mucosal FA composition, resulting in increased linoleic acid (11.52-16.33% of total FAs) and ARA (2.35-5.16% of total FAs). CONCLUSIONS Prebiotic supplementation during the first 22 d of life altered the gut microbiota of piglets and increased the abundance of specific bacterial genera. These changes correlated with increased SCFA, which may benefit intestinal development. Although dietary ARA did not alter the microbiota, it increased the ARA content of the colonic mucosa, which may support intestinal development and epithelial repair. Prebiotic supplementation also increased unsaturation of FAs in the colonic mucosa. Although the mechanism requires further investigation, it may be related to altered microbial ecology or biohydrogenation of FA.
Collapse
Affiliation(s)
- Brandon J Eudy
- Department of Animal Science and Laboratory of Developmental Nutrition, North Carolina State University, Raleigh, NC, United States
| | - Jack Odle
- Department of Animal Science and Laboratory of Developmental Nutrition, North Carolina State University, Raleigh, NC, United States.
| | - Xi Lin
- Department of Animal Science and Laboratory of Developmental Nutrition, North Carolina State University, Raleigh, NC, United States
| | - Christian Maltecca
- Department of Animal Science and Laboratory of Developmental Nutrition, North Carolina State University, Raleigh, NC, United States
| | - Kathleen R Walter
- Department of Animal Science and Laboratory of Developmental Nutrition, North Carolina State University, Raleigh, NC, United States
| | - Nathan P McNulty
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, United States
| | - Vivek Fellner
- Department of Animal Science and Laboratory of Developmental Nutrition, North Carolina State University, Raleigh, NC, United States
| | - Sheila K Jacobi
- Department of Animal Sciences, The Ohio State University, Columbus, OH, United States.
| |
Collapse
|
8
|
Zeng X, Li S, Liu L, Cai S, Ye Q, Xue B, Wang X, Zhang S, Chen F, Cai C, Wang F, Zeng X. Role of functional fatty acids in modulation of reproductive potential in livestock. J Anim Sci Biotechnol 2023; 14:24. [PMID: 36788613 PMCID: PMC9926833 DOI: 10.1186/s40104-022-00818-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/04/2022] [Indexed: 02/16/2023] Open
Abstract
Fatty acids are not only widely known as energy sources, but also play important roles in many metabolic pathways. The significance of fatty acids in modulating the reproductive potential of livestock has received greater recognition in recent years. Functional fatty acids and their metabolites improve follicular development, oocyte maturation and embryo development, as well as endometrial receptivity and placental vascular development, through enhancing energy supply and precursors for the synthesis of their productive hormones, such as steroid hormones and prostaglandins. However, many studies are focused on the impacts of individual functional fatty acids in the reproductive cycle, lacking studies involved in deeper mechanisms and optimal fatty acid requirements for specific physiological stages. Therefore, an overall consideration of the combination and synergy of functional fatty acids and the establishment of optimal fatty acid requirement for specific stages is needed to improve reproductive potential in livestock.
Collapse
Affiliation(s)
- Xiangzhou Zeng
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, 100193 Beijing, P. R. China ,Beijing Key Laboratory of Bio feed Additives, 100193 Beijing, P. R. China
| | - Siyu Li
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, 100193 Beijing, P. R. China ,Beijing Key Laboratory of Bio feed Additives, 100193 Beijing, P. R. China
| | - Lu Liu
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, 100193 Beijing, P. R. China ,Beijing Key Laboratory of Bio feed Additives, 100193 Beijing, P. R. China
| | - Shuang Cai
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, 100193 Beijing, P. R. China ,Beijing Key Laboratory of Bio feed Additives, 100193 Beijing, P. R. China
| | - Qianhong Ye
- grid.35155.370000 0004 1790 4137State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, 430070 Wuhan, Hubei China
| | - Bangxin Xue
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, 100193 Beijing, P. R. China ,Beijing Key Laboratory of Bio feed Additives, 100193 Beijing, P. R. China
| | - Xinyu Wang
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, 100193 Beijing, P. R. China ,Beijing Key Laboratory of Bio feed Additives, 100193 Beijing, P. R. China
| | - Shihai Zhang
- grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Fang Chen
- grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Chuanjiang Cai
- grid.144022.10000 0004 1760 4150College of Animal Science and Technology, Northwest A&F University, 712100 Yangling, Shaanxi China
| | - Fenglai Wang
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, 100193 Beijing, P. R. China ,Beijing Key Laboratory of Bio feed Additives, 100193 Beijing, P. R. China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, 100193, Beijing, P. R. China. .,Beijing Key Laboratory of Bio feed Additives, 100193, Beijing, P. R. China.
| |
Collapse
|
9
|
Xiao K, Yang Y, Zhang Y, Lv Q, Huang F, Wang D, Zhao J, Liu Y. Long-chain PUFA ameliorate enterotoxigenic Escherichia coli-induced intestinal inflammation and cell injury by modulating pyroptosis and necroptosis signaling pathways in porcine intestinal epithelial cells. Br J Nutr 2022; 128:835-850. [PMID: 34915950 DOI: 10.1017/s0007114521005092] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This study was aimed to investigate whether EPA and arachidonic acid (ARA), the representative n-3 or n-6 PUFA, could alleviate enterotoxigenic Escherichia coli (ETEC) K88-induced inflammation and injury of intestinal porcine epithelial cells 1 (IPEC-1) by modulating pyroptosis and necroptosis signalling pathways. IPEC-1 cells were cultured with or without EPA or ARA in the presence or absence of ETEC K88. EPA and ARA reduced ETEC K88 adhesion and endotoxin content in the supernatant. EPA and ARA increased transepithelial electrical resistance, decreased permeability of fluorescein isothiocyanate-labelled dextran, increased membrane protein expression of occludin, ZO-1 and claudin-1 and relieved disturbed distribution of these proteins. EPA and ARA also reduced cell necrosis ratio. EPA or ARA reduced mRNA and concentration of TNF-α, IL-6 and IL-8 and decreased mRNA abundances of intestinal toll-like receptors 4 and its downstream signals. Moreover, EPA and ARA downregulated mRNA expression of nod-like receptor protein 3 (NLRP3), caspase 1 and IL-18 and inhibited protein expression of NLRP3, apoptosis-associated speck-like protein containing a CARD (ASC), gasdermin D and caspase-1. Finally, EPA and ARA reduced mRNA expression of fas-associated death domain protein, caspase 8, receptor-interacting protein kinase (RIP) 1, mixed lineage kinase-like protein (MLKL), phosphoglycerate mutase 5 (PGAM5), motility-related protein 1 (Drp1) and high mobility protein 1 (HMGB1) and inhibited protein expression of phosphorylated-RIP1, p-RIP3, p-MLKL and HMGB1. These data demonstrate that EPA and ARA prevent ETEC K88-induced cell inflammation and injury, which is partly through inhibiting pyroptosis and necroptosis signalling pathways.
Collapse
Affiliation(s)
- Kan Xiao
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Yang Yang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Yang Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Qingqing Lv
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Feifei Huang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Dan Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Jiangchao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR, USA
| | - Yulan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| |
Collapse
|
10
|
Sundaram TS, Giromini C, Rebucci R, Pistl J, Bhide M, Baldi A. Role of omega-3 polyunsaturated fatty acids, citrus pectin, and milk-derived exosomes on intestinal barrier integrity and immunity in animals. J Anim Sci Biotechnol 2022; 13:40. [PMID: 35399093 PMCID: PMC8996583 DOI: 10.1186/s40104-022-00690-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/07/2022] [Indexed: 11/10/2022] Open
Abstract
The gastrointestinal tract of livestock and poultry is prone to challenge by feedborne antigens, pathogens, and other stress factors in the farm environment. Excessive physiological inflammation and oxidative stress that arises firstly disrupts the intestinal epithelial barrier followed by other components of the gastrointestinal tract. In the present review, the interrelationship between intestinal barrier inflammation and oxidative stress that contributes to the pathogenesis of inflammatory bowel disease was described. Further, the role of naturally existing immunomodulatory nutrients such as the omega-3 polyunsaturated fatty acids, citrus pectin, and milk-derived exosomes in preventing intestinal barrier inflammation was discussed. Based on the existing evidence, the possible molecular mechanism of these bioactive nutrients in the intestinal barrier was outlined for application in animal diets.
Collapse
Affiliation(s)
- Tamil Selvi Sundaram
- Department of Veterinary Science for Health, Animal Production and Food Safety, University of Milan, Via Trentacoste 2, 20134, Milan, Italy.
- University of Veterinary Medicine and Pharmacy in Košice, Komenského 68/73, 04181, Košice, Slovakia.
| | - Carlotta Giromini
- Department of Veterinary Science for Health, Animal Production and Food Safety, University of Milan, Via Trentacoste 2, 20134, Milan, Italy
| | - Raffaella Rebucci
- Department of Veterinary Science for Health, Animal Production and Food Safety, University of Milan, Via Trentacoste 2, 20134, Milan, Italy
| | - Juraj Pistl
- University of Veterinary Medicine and Pharmacy in Košice, Komenského 68/73, 04181, Košice, Slovakia
| | - Mangesh Bhide
- University of Veterinary Medicine and Pharmacy in Košice, Komenského 68/73, 04181, Košice, Slovakia
| | - Antonella Baldi
- Department of Veterinary Science for Health, Animal Production and Food Safety, University of Milan, Via Trentacoste 2, 20134, Milan, Italy
| |
Collapse
|
11
|
Cioce M, Canino C, Pass H, Blandino G, Strano S, Fazio VM. Arachidonic acid drives adaptive responses to chemotherapy-induced stress in malignant mesothelioma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:344. [PMID: 34727953 PMCID: PMC8561918 DOI: 10.1186/s13046-021-02118-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/24/2021] [Indexed: 12/13/2022]
Abstract
Background High resistance to therapy and poor prognosis characterizes malignant pleural mesothelioma (MPM). In fact, the current lines of treatment, based on platinum and pemetrexed, have limited impact on the survival of MPM patients. Adaptive response to therapy-induced stress involves complex rearrangements of the MPM secretome, mediated by the acquisition of a senescence-associated-secretory-phenotype (SASP). This fuels the emergence of chemoresistant cell subpopulations, with specific gene expression traits and protumorigenic features. The SASP-driven rearrangement of MPM secretome takes days to weeks to occur. Thus, we have searched for early mediators of such adaptive process and focused on metabolites differentially released in mesothelioma vs mesothelial cell culture media, after treatment with pemetrexed. METHODS Mass spectrometry-based (LC/MS and GC/MS) identification of extracellular metabolites and unbiased statistical analysis were performed on the spent media of mesothelial and mesothelioma cell lines, at steady state and after a pulse with pharmacologically relevant doses of the drug. ELISA based evaluation of arachidonic acid (AA) levels and enzyme inhibition assays were used to explore the role of cPLA2 in AA release and that of LOX/COX-mediated processing of AA. QRT-PCR, flow cytometry analysis of ALDH expressing cells and 3D spheroid growth assays were employed to assess the role of AA at mediating chemoresistance features of MPM. ELISA based detection of p65 and IkBalpha were used to interrogate the NFkB pathway activation in AA-treated cells. RESULTS We first validated what is known or expected from the mechanism of action of the antifolate. Further, we found increased levels of PUFAs and, more specifically, arachidonic acid (AA), in the transformed cell lines treated with pemetrexed. We showed that pharmacologically relevant doses of AA tightly recapitulated the rearrangement of cell subpopulations and the gene expression changes happening in pemetrexed -treated cultures and related to chemoresistance. Further, we showed that release of AA following pemetrexed treatment was due to cPLA2 and that AA signaling impinged on NFkB activation and largely affected anchorage-independent, 3D growth and the resistance of the MPM 3D cultures to the drug. CONCLUSIONS AA is an early mediator of the adaptive response to pem in chemoresistant MPM and, possibly, other malignancies.
Collapse
Affiliation(s)
- Mario Cioce
- Department of Medicine, R.U. in Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, 00128, Rome, Italy.
| | - Claudia Canino
- Division of General Thoracic Surgery, Department of Cardiothoracic Surgery, NYU Langone Medical Center, New York, NY, USA.,Radiation Oncology Unit, UPMC Hillmann Cancer Center, San Pietro Hospital FBF, Rome, Italy
| | - Harvey Pass
- Division of General Thoracic Surgery, Department of Cardiothoracic Surgery, NYU Langone Medical Center, New York, NY, USA
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Sabrina Strano
- SAFU Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Vito Michele Fazio
- Department of Medicine, R.U. in Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, 00128, Rome, Italy. .,Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00133, Rome, Italy. .,Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo, Italy.
| |
Collapse
|
12
|
Yu J, Zhang W, Chi X, Chen W, Li Z, Wang Y, Liu Z, Wang H, Xu B. The dietary arachidonic acid improved growth and immunity of honey bee ( Apis mellifera ligustica). BULLETIN OF ENTOMOLOGICAL RESEARCH 2021; 112:1-10. [PMID: 34622750 DOI: 10.1017/s0007485321000821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Honeybees cannot synthesize arachidonic acid (ARA) themselves, only obtain it from food. Most pollen is deficient or contains a small amount of ARA. The necessity of supplementary ARA in bees' diet has not been studied. The objective of this study was to investigate the effects of dietary ARA levels on the growth and immunity of Apis mellifera ligustica. A total of 25 honeybee colonies were randomly assigned to five dietary groups which were fed basic diets supplemented with 0, 2, 4, 6, and 8% of ARA. The diet with 4% ARA improved the body weight of newly emerged worker bees compared with the control group. Supplement of ARA in honeybee diets changed the fatty acid composition of honeybee body. SFA and MUFA contents of bees' body declined, and PUFA content rised in the ARA group. Compared with the control group, the supplement of ARA in honeybee diets increased the contents of ARA, C22:6n-3 (DHA) and C18:3n-6 in bees' body significantly, but decreased the contents of C16:1 and C18:3n-3. The diet supplied with 4% ARA reduced the mortality rate of honeybee infected with Escherichia coli. The activity of immune enzymes (phenoloxidase, antitrypsin, and lysozyme) and the mRNA expression levels of immune genes (defensin-2, toll, myd88, and dorsal) were improved by ARA diets to varying degrees depending on the ARA levels, especially 4% ARA. These results suggested that dietary ARA could improve the growth, survival, and immune functions of honeybees. Supplement of ARA in bees' diet would be valuable for the fitness of honeybees.
Collapse
Affiliation(s)
- Jing Yu
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China
| | - Weixing Zhang
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China
| | - Xuepeng Chi
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China
| | - Wenfeng Chen
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China
| | - Zhenfang Li
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China
| | - Ying Wang
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China
| | - Zhenguo Liu
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China
| | - Hongfang Wang
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China
| | - Baohua Xu
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China
| |
Collapse
|
13
|
Meng F, Qiu J, Chen H, Shi X, Yin M, Zhu M, Yang G. Dietary supplementation with N-3 polyunsaturated fatty acid-enriched fish oil promotes wound healing after ultraviolet B-induced sunburn in mice. Food Sci Nutr 2021; 9:3693-3700. [PMID: 34262728 PMCID: PMC8269668 DOI: 10.1002/fsn3.2330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 03/25/2021] [Accepted: 04/25/2021] [Indexed: 12/14/2022] Open
Abstract
N-3 polyunsaturated fatty acids (n-3 PUFA) can alleviate ultraviolet B (UVB)-induced skin cancers, but their effects on sunburn and upcoming wound healing remain controversial. This study aimed to explore the impact of n-3 PUFA-enriched fish oil (n-3 PUFA-FO) on UVB-induced sunburns and subsequent healing. Sixty C57BL/6 female mice were divided into two groups. The treated group mice were fed n-3 PUFA-FO for the entire duration of the experiment. Mice in the control group were fed a standard diet. After two weeks of n-3 PUFA-FO feeding, mice were exposed to UVB for 20 min and sacrificed 20 d later. Skin photodamage and lesion area were recorded during wound healing. Epidermal lesion thickness was quantified in hematoxylin and eosin-stained skin sections. Inflammation and macrophage polarization were assessed by qRT-PCR. Oxidative stress and antioxidant enzyme activity were quantified using specific ELISA kits. N-3 PUFA-FO feeding decreased UVB photodamage and accelerated wound healing progression, both of which were coupled with less intense inflammation and increased macrophage M2 phenotype polarization. Furthermore, n-3 PUFA-FO brought about a decrease in malondialdehyde (MDA) levels but increased the activity of catalase (CAT) and glutathione peroxidase (GP), without changing superoxide dismutase (SOD) activity. N-3 PUFA-FO protects against UVB-induced skin photodamage and promotes wound healing by modulating macrophage phenotypic polarization and antioxidant enzyme activities. N-3 PUFA-FO could be a novel therapeutic approach for both the prevention and treatment of sunburns.
Collapse
Affiliation(s)
- Fanxing Meng
- College of Animal Science and TechnologyJilin Agricultural UniversityChangchunChina
| | | | - Houjie Chen
- The Shenzhen Key Laboratory of Health Sciences and TechnologyInternational Graduate School at ShenzhenTsinghua UniversityShenzhenChina
| | - Xiaojun Shi
- The Shenzhen Key Laboratory of Health Sciences and TechnologyInternational Graduate School at ShenzhenTsinghua UniversityShenzhenChina
| | - Meifang Yin
- Department of Burn and Plastic SurgeryDepartment of Wound RepairShenzhen Institute of Translational MedicineThe First Affiliated Hospital of Shenzhen University Health Science CenterShenzhen Second People's HospitalShenzhenChina
| | - Meishu Zhu
- Department of Burn and Plastic SurgeryDepartment of Wound RepairShenzhen Institute of Translational MedicineThe First Affiliated Hospital of Shenzhen University Health Science CenterShenzhen Second People's HospitalShenzhenChina
| | - Guang Yang
- Department of Burn and Plastic SurgeryDepartment of Wound RepairShenzhen Institute of Translational MedicineThe First Affiliated Hospital of Shenzhen University Health Science CenterShenzhen Second People's HospitalShenzhenChina
| |
Collapse
|
14
|
Huang N, Wang M, Peng J, Wei H. Role of arachidonic acid-derived eicosanoids in intestinal innate immunity. Crit Rev Food Sci Nutr 2020; 61:2399-2410. [PMID: 32662287 DOI: 10.1080/10408398.2020.1777932] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Arachidonic acid (ARA), an n-6 essential fatty acid, plays an important role in human and animal growth and development. The ARA presents in the membrane phospholipids can be released by phospholipase A2. These free arachidonic acid molecules are then used to produce eicosanoids through three different pathways. Previous studies have demonstrated that eicosanoids have a wide range of physiological functions. Although they are generally considered to be pro-inflammatory molecules, recent advances have elucidated they have an effect on innate immunity via regulating the development, and differentiation of innate immune cells and the function of the intestinal epithelial barrier. Here, we review eicosanoids generation in intestine and their role in intestinal innate immunity, focusing on intestinal epithelial barrier, innate immune cell in lamina propria (LP) and their crosstalk.
Collapse
Affiliation(s)
- Ningning Huang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Miaomiao Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| |
Collapse
|
15
|
Li Z, Xu B, Lu Z, Wang Y. Effects of long-chain fatty acid supplementation on the growth performance of grower and finisher pigs: a meta-analysis. J Anim Sci Biotechnol 2019; 10:65. [PMID: 31428366 PMCID: PMC6696677 DOI: 10.1186/s40104-019-0374-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 06/21/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Supplementation of feed with long-chain fatty acids (LCFAs) during the grower and finisher phases has long been discussed as a growth promotion strategy in pigs, but its effects are inconsistent. The purpose of our study was to comprehensively evaluate its effects on the growth performance based on the average daily gain (ADG), average daily feed intake (ADFI) and gain: feed (G:F) ratio and to unveil the roles of the basal diet, LCFA concentration and LCFA saturation. RESULTS We searched the PubMed and Web of Science databases (articles published from Jan 1st, 2000, to Sep 30th, 2018; restricted to English) and compared LCFA-supplemented diets with control diets. We retrieved 2346 studies, 18 of which (1314 pigs, 26 records) were eligible for our analysis. We used a random-effects model to calculate the weighted mean differences (WMDs) and 95% confidence intervals (CIs). LCFA supplementation in the grower-finisher phase improved the ADG (WMD = 41.74 g/d, 95% CI: 8.81 to 74.66, P = 0.013) and G:F ratio (WMD = 0.019, 95% CI: 0.006 to 0.032, P = 0.003). For supplementation solely in the finisher phase, we found a similar performance in the ADG (WMD = 39.93 g/d, 95% CI: 26.48 to 53.38, P < 0.001) and G:F ratio (WMD = 0.019, 95% CI: 0.006 to 0.032, P < 0.001) but a reduction in the ADFI (WMD = - 83.863 g/d, 95% CI: - 156.157 to - 11.569, P = 0.023). Specifically, approximately 5% LCFA supplementation in the finisher phase had significant effects on the ADG (WMD = 51.385 g/d, 95% CI: 35.816 to 66.954, P < 0.001), ADFI (WMD = - 102.869 g/d, 95% CI: - 189.236 to - 16.502, P = 0.02) and G:F ratio (WMD = 0.028, 95% CI: 0.018 to 0.039, P < 0.001), whereas a concentration of approximately 1% exhibited no effects. CONCLUSIONS Overall, regardless of the basal diet and saturation, LCFA supplementation greatly improves the growth performance of grower and finisher pigs, primarily by increasing the energy density.
Collapse
Affiliation(s)
- Zhi Li
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed of Ministry of Agriculture, Key Laboratory of Animal Nutrition and Feed Science of Zhejiang Province, Institute of Feed Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang Province 310058 People’s Republic of China
| | - Bocheng Xu
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed of Ministry of Agriculture, Key Laboratory of Animal Nutrition and Feed Science of Zhejiang Province, Institute of Feed Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang Province 310058 People’s Republic of China
| | - Zeqing Lu
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed of Ministry of Agriculture, Key Laboratory of Animal Nutrition and Feed Science of Zhejiang Province, Institute of Feed Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang Province 310058 People’s Republic of China
| | - Yizhen Wang
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed of Ministry of Agriculture, Key Laboratory of Animal Nutrition and Feed Science of Zhejiang Province, Institute of Feed Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang Province 310058 People’s Republic of China
| |
Collapse
|
16
|
Walter KR, Lin X, Jacobi SK, Käser T, Esposito D, Odle J. Dietary arachidonate in milk replacer triggers dual benefits of PGE 2 signaling in LPS-challenged piglet alveolar macrophages. J Anim Sci Biotechnol 2019; 10:13. [PMID: 30815256 PMCID: PMC6376662 DOI: 10.1186/s40104-019-0321-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/09/2019] [Indexed: 12/23/2022] Open
Abstract
Background Respiratory infections challenge the swine industry, despite common medicinal practices. The dual signaling nature of PGE2 (supporting both inflammation and resolution) makes it a potent regulator of immune cell function. Therefore, the use of dietary long chain n-6 PUFA to enhance PGE2 effects merits investigation. Methods Day-old pigs (n = 60) were allotted to one of three dietary groups for 21 d (n = 20/diet), and received either a control diet (CON, arachidonate = 0.5% of total fatty acids), an arachidonate (ARA)-enriched diet (LC n-6, ARA = 2.2%), or an eicosapentaenoic (EPA)-enriched diet (LC n-3, EPA = 3.0%). Alveolar macrophages and lung parenchymal tissue were collected for fatty acid analysis. Isolated alveolar macrophages were stimulated with LPS in situ for 24 h, and mRNA was isolated to assess markers associated with inflammation and eicosanoid production. Culture media were collected to assess PGE2 secretion. Oxidative burst in macrophages was measured by: 1) oxygen consumption and extracellular acidification (via Seahorse), 2) cytoplasmic oxidation and 3) nitric oxide production following 4, 18, and 24 h of LPS stimulation. Results Concentration of ARA (% of fatty acids, w/w) in macrophages from pigs fed LC n-6 was 86% higher than CON and 18% lower in pigs fed LC n-3 (P < 0.01). Following LPS stimulation, abundance of COX-2 and TNF-α mRNA (P < 0.0001), and PGE2 secretion (P < 0. 01) were higher in LC n-6 PAM vs. CON. However, ALOX5 abundance was 1.6-fold lower than CON. Macrophages from CON and LC n-6 groups were 4-fold higher in ALOX12/15 abundance (P < 0.0001) compared to LC n-3. Oxygen consumption and extracellular acidification rates increased over 4 h following LPS stimulation (P < 0.05) regardless of treatment. Similarly, increases in cytoplasmic oxidation (P < 0.001) and nitric oxide production (P < 0.002) were observed after 18 h of LPS stimulation but were unaffected by diet. Conclusions We infer that enriching diets with arachidonic acid may be an effective means to enhance a stronger innate immunologic response to respiratory challenges in neonatal pigs. However, further work is needed to examine long-term safety, clinical efficacy and economic viability. Electronic supplementary material The online version of this article (10.1186/s40104-019-0321-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kathleen R Walter
- 1Department of Animal Science, Plants for Human Health Institute, North Carolina State University, Kannapolis, North Carolina USA.,2Department of Animal Science, North Carolina State University, Raleigh, North Carolina USA
| | - Xi Lin
- 2Department of Animal Science, North Carolina State University, Raleigh, North Carolina USA
| | - Sheila K Jacobi
- 3Department of Animal Science, Ohio State University, Columbus, Ohio USA
| | - Tobias Käser
- 4Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina USA
| | - Debora Esposito
- 1Department of Animal Science, Plants for Human Health Institute, North Carolina State University, Kannapolis, North Carolina USA.,2Department of Animal Science, North Carolina State University, Raleigh, North Carolina USA
| | - Jack Odle
- 2Department of Animal Science, North Carolina State University, Raleigh, North Carolina USA
| |
Collapse
|
17
|
Ziegler AL, Pridgen TA, Mills JK, Gonzalez LM, Van Landeghem L, Odle J, Blikslager AT. Epithelial restitution defect in neonatal jejunum is rescued by juvenile mucosal homogenate in a pig model of intestinal ischemic injury and repair. PLoS One 2018; 13:e0200674. [PMID: 30138372 PMCID: PMC6107120 DOI: 10.1371/journal.pone.0200674] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 08/02/2018] [Indexed: 12/16/2022] Open
Abstract
Intestinal ischemic injury results sloughing of the mucosal epithelium leading to host sepsis and death unless the mucosal barrier is rapidly restored. Volvulus and neonatal necrotizing enterocolitis (NEC) in infants have been associated with intestinal ischemia, sepsis and high mortality rates. We have characterized intestinal ischemia/repair using a highly translatable porcine model in which juvenile (6-8-week-old) pigs completely and efficiently restore barrier function by way of rapid epithelial restitution and tight junction re-assembly. In contrast, separate studies showed that younger neonatal (2-week-old) pigs exhibited less robust recovery of barrier function, which may model an important cause of high mortality rates in human infants with ischemic intestinal disease. Therefore, we aimed to further refine our repair model and characterize defects in neonatal barrier repair. Here we examine the defect in neonatal mucosal repair that we hypothesize is associated with hypomaturity of the epithelial and subepithelial compartments. Following jejunal ischemia in neonatal and juvenile pigs, injured mucosa was stripped from seromuscular layers and recovered ex vivo while monitoring transepithelial electrical resistance (TEER) and 3H-mannitol flux as measures of barrier function. While ischemia-injured juvenile mucosa restored TEER above control levels, reduced flux over the recovery period and showed 93±4.7% wound closure, neonates exhibited no change in TEER, increased flux, and a 11±23.3% increase in epithelial wound size. Scanning electron microscopy revealed enterocytes at the wound margins of neonates failed to assume the restituting phenotype seen in restituting enterocytes of juveniles. To attempt rescue of injured neonatal mucosa, neonatal experiments were repeated with the addition of exogenous prostaglandins during ex vivo recovery, ex vivo recovery with full thickness intestine, in vivo recovery and direct application of injured mucosal homogenate from neonates or juveniles. Neither exogenous prostaglandins, intact seromuscular intestinal layers, nor in vivo recovery enhanced TEER or restitution in ischemia-injured neonatal mucosa. However, ex vivo exogenous application of injured juvenile mucosal homogenate produced a significant increase in TEER and enhanced histological restitution to 80±4.4% epithelial coverage in injured neonatal mucosa. Thus, neonatal mucosal repair can be rescued through direct contact with the cellular and non-cellular milieu of ischemia-injured mucosa from juvenile pigs. These findings support the hypothesis that a defect in mucosal repair in neonates is due to immature repair mechanisms within the mucosal compartment. Future studies to identify and rescue specific defects in neonatal intestinal repair mechanisms will drive development of novel clinical interventions to reduce mortality in infants affected by intestinal ischemic injury.
Collapse
Affiliation(s)
- Amanda L. Ziegler
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Tiffany A. Pridgen
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Juliana K. Mills
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Liara M. Gonzalez
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Laurianne Van Landeghem
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Jack Odle
- Department of Animal Science, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Anthony T. Blikslager
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
18
|
Wound Healing and Omega-6 Fatty Acids: From Inflammation to Repair. Mediators Inflamm 2018; 2018:2503950. [PMID: 29849484 PMCID: PMC5925018 DOI: 10.1155/2018/2503950] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/08/2018] [Indexed: 12/25/2022] Open
Abstract
Wound healing is an evolutionarily conserved process that is essential for species survival. Wound healing involves a series of biochemical and cellular events that are tightly controlled, divided into 3 concomitant and overlapping phases: inflammation, proliferation, and remodelling. Poor wound healing or a chronic wound represents a silent epidemic that affects billions of people worldwide. Considering the involvement of immune cells in its resolution, recent studies are focused on investigating the roles of immune nutrients such as amino acids, minerals, and fatty acids on wound healing. Among the fatty acids, much attention has been given to omega-6 (ω-6) fatty acids since they can modulate cell migration and proliferation, phagocytic capacity, and production of inflammatory mediators. The present review summarizes current knowledge about the role of ω-6 fatty acids in the wound healing context.
Collapse
|
19
|
Schuck-Phan A, Phan T, Dawson PA, Dial EJ, Bell C, Liu Y, Rhoads JM, Lichtenberger LM. Formula Feeding Predisposes Gut to NSAID-Induced Small Intestinal Injury. ACTA ACUST UNITED AC 2016; 6. [PMID: 31565540 DOI: 10.4172/2161-1459.1000222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Objectives Breast feeding protects infants from many diseases, including necrotizing enterocolitis, peptic ulceration and infectious diarrhea. Conversely, maternal separation stress and Non-Steroidal Anti-Inflammatory Drugs (NSAID's) can induce intestinal injury and bleeding. This study aimed to evaluate in suckling rats if maternal separation/formula feeding leads to increased intestinal sensitivity to indomethacin (indo)-induced intestinal injury and to look at potential mechanisms involved. Methods Nine-day-old rats were dam-fed or separated/trained to formula-feed for 6 days prior to indo administration (5 mg/kg/day) or saline (control) for 3 days. Intestinal bleeding and injury were assessed by measuring luminal and Fecal Hemoglobin (Hob) and jejunal histology. Maturation of the intestine was assessed by measuring luminal bile acids, jejunal sucrase, serum corticosterone, and mRNA expression of ileal Apical Sodium-Dependent Bile Acid Transporter (ASBT). Results At 17 days, formula-fed indo-treated pups had a 2-fold increase in luminal Hb compared to formula-fed control pups and had evidence of morphological injury to the small intestinal mucosa as observed at the light microscopic level, whereas indo had no effect on dam-fed littermates. In addition, formula-fed rats had significant increases in luminal bile acid, sucrase specific activity, serum corticosterone, and expression of ASBT mRNA compared to dam-fed rats. Conclusion Maternal separation stress may cause early intestinal maturational changes induced by corticosteroid release, including increased epithelial exposure to bile acids. These maturational changes may have a sensitizing rather than protective effect against indo-induced injury in the new-born.
Collapse
Affiliation(s)
- A Schuck-Phan
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Texas Health Science Center, Houston, TX, USA
| | - T Phan
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
| | - P A Dawson
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - E J Dial
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
| | - C Bell
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Texas Health Science Center, Houston, TX, USA
| | - Y Liu
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Texas Health Science Center, Houston, TX, USA
| | - J M Rhoads
- Department of Pediatrics, Division of Pediatric Gastroenterology, University of Texas Health Science Center, Houston, TX, USA
| | - L M Lichtenberger
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|
20
|
Abstract
Gastrointestinal disease is a prevalent cause of morbidity and mortality and the use of animal models have been instrumental in studying mechanisms of digestive pathophysiology. As investigators attempt to translate the wealth of basic science information developed from rodent, models, large animal models provide a number of translational advantages. The pig, in particular, is arguably one of the most powerful models of human organ systems, including the gastrointestinal tract. The pig has provided important tools and insight into intestinal ischemia/reperfusion injury, intestinal mucosal repair, as well as new insights into esophageal injury and repair. Porcine model development has taken advantage of the size of the animal, allowing increased surgical and endoscopic access. In addition, cellular tools such as the intestinal porcine epithelial cell line and porcine enteroids are providing the methodology to translate basic science findings using in-depth mechanistic analyses. Further opportunities in porcine digestive disease modeling include developing additional transgenic pig strains. Collectively, porcine models hold great promise for the future of clinically relevant digestive disease research.
Collapse
|
21
|
Glover LE, Lee JS, Colgan SP. Oxygen metabolism and barrier regulation in the intestinal mucosa. J Clin Invest 2016; 126:3680-3688. [PMID: 27500494 DOI: 10.1172/jci84429] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mucosal surfaces are lined by epithelial cells and provide an important barrier to the flux of antigens from the outside. This barrier is provided at a number of levels, including epithelial junctional complexes, mucus production, and mucosa-derived antimicrobials. Tissue metabolism is central to the maintenance of homeostasis in the mucosa. In the intestine, for example, baseline pO2 levels are uniquely low due to counter-current blood flow and the presence of large numbers of bacteria. As such, hypoxia and HIF signaling predominates normal intestinal metabolism and barrier regulation during both homeostasis and active inflammation. Contributing factors that elicit important adaptive responses within the mucosa include the transcriptional regulation of tight junction proteins, metabolic regulation of barrier components, and changes in autophagic flux. Here, we review recent literature around the topic of hypoxia and barrier function in health and during disease.
Collapse
|
22
|
Rosero DS, Boyd RD, Odle J, van Heugten E. Optimizing dietary lipid use to improve essential fatty acid status and reproductive performance of the modern lactating sow: a review. J Anim Sci Biotechnol 2016; 7:34. [PMID: 27274395 PMCID: PMC4895830 DOI: 10.1186/s40104-016-0092-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 05/13/2016] [Indexed: 11/24/2022] Open
Abstract
Dietary lipid supplementation benefits the prolific and high-producing modern lactating sow. A comprehensive review of recent studies showed that lipid supplementation increases average daily energy intake, which is partitioned for lactation as indicated by greater milk fat output and improved litter growth rate. Recent compelling findings showed that addition of particular lipids during lactation improved the subsequent reproductive outcome of sows. Such benefits were related to the level of dietary essential fatty acids (EFA, linoleic acid, C18:2n-6; and α-linolenic acid, C18:3n-3) during lactation. Lactation diets without supplemental EFA resulted in a pronounced negative balance (intake minus milk output) of linoleic (−25.49 g/d) and α-linolenic acid (−2.75 g/d); which compromised sow fertility (farrowing rate < 75 % and culling rates > 25 % of weaned sows). This phenomenon seems to be increasingly important with advancing sow age because of a progressive reduction of body EFA pool over successive lactations. The net effect of supplemental EFA during lactation was to create a positive EFA balance, which improved the subsequent reproduction of sows. Adequate linoleic acid intake improved the proportion of sows that farrowed in the subsequent cycle (Farrowing rate (%) = [(−1.5 × 10−3 × linoleic acid intake (g/d)2) + (0.53 × linoleic acid intake (g/d)) + (45.2)]; quadratic P = 0.002, R2 = 0.997, RMSE = 0.031). In addition, increasing linoleic acid intake increased the number of pigs born in the subsequent cycle (total pigs born (n) = [(9.4 × 10−5 × linoleic acid intake (g/d)2) + (0.04 × linoleic acid intake (g/d)) + (10.94)]; quadratic P = 0.002, R2 = 0.997, RMSE = 0.031). Supplemental α-linolenic acid resulted in a rapid return to estrus (sows bred: sows weaned = 94.2 %; wean-to-estrus interval = 4.0 d) and achieved a high retention of pregnancy (sows pregnant: sows bred = 98 %). Collectively, we conclude that a minimum dietary intake of 10 g/d of α-linolenic acid, simultaneous with a minimum of 125 g/d of linoleic acid should be provided to ≥ 95 % of the sows; thereby, achieving a maximum sow reproductive efficiency through multiple mechanisms that include rapid return to estrus, high maintenance of pregnancy and large subsequent litter size in mature sows, that appear to be susceptible to EFA deficiency.
Collapse
Affiliation(s)
| | - R Dean Boyd
- The Hanor Company, Franklin, KY 42134 USA ; Department of Animal Science, North Carolina State University, Raleigh, NC 27695 USA
| | - Jack Odle
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695 USA
| | - Eric van Heugten
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695 USA
| |
Collapse
|
23
|
Hadley KB, Ryan AS, Forsyth S, Gautier S, Salem N. The Essentiality of Arachidonic Acid in Infant Development. Nutrients 2016; 8:216. [PMID: 27077882 PMCID: PMC4848685 DOI: 10.3390/nu8040216] [Citation(s) in RCA: 241] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/01/2016] [Accepted: 04/05/2016] [Indexed: 01/16/2023] Open
Abstract
Arachidonic acid (ARA, 20:4n-6) is an n-6 polyunsaturated 20-carbon fatty acid formed by the biosynthesis from linoleic acid (LA, 18:2n-6). This review considers the essential role that ARA plays in infant development. ARA is always present in human milk at a relatively fixed level and is accumulated in tissues throughout the body where it serves several important functions. Without the provision of preformed ARA in human milk or infant formula the growing infant cannot maintain ARA levels from synthetic pathways alone that are sufficient to meet metabolic demand. During late infancy and early childhood the amount of dietary ARA provided by solid foods is low. ARA serves as a precursor to leukotrienes, prostaglandins, and thromboxanes, collectively known as eicosanoids which are important for immunity and immune response. There is strong evidence based on animal and human studies that ARA is critical for infant growth, brain development, and health. These studies also demonstrate the importance of balancing the amounts of ARA and DHA as too much DHA may suppress the benefits provided by ARA. Both ARA and DHA have been added to infant formulas and follow-on formulas for more than two decades. The amounts and ratios of ARA and DHA needed in infant formula are discussed based on an in depth review of the available scientific evidence.
Collapse
Affiliation(s)
- Kevin B Hadley
- DSM Nutritional Products, 6480 Dobbin Road, Columbia, MD 21045, USA.
| | - Alan S Ryan
- Clinical Research Consulting, 9809 Halston Manor, Boynton Beach, FL 33473, USA.
| | - Stewart Forsyth
- School of Medicine, Dentistry & Nursing, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK.
| | - Sheila Gautier
- DSM Nutritional Products, 6480 Dobbin Road, Columbia, MD 21045, USA.
| | - Norman Salem
- DSM Nutritional Products, 6480 Dobbin Road, Columbia, MD 21045, USA.
| |
Collapse
|
24
|
Rosero DS, Boyd RD, McCulley M, Odle J, van Heugten E. Essential fatty acid supplementation during lactation is required to maximize the subsequent reproductive performance of the modern sow. Anim Reprod Sci 2016; 168:151-163. [PMID: 27037065 DOI: 10.1016/j.anireprosci.2016.03.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 03/23/2016] [Accepted: 03/25/2016] [Indexed: 10/22/2022]
Abstract
This study was conducted to investigate the effects of supplemental essential fatty acids (EFA) on sow reproductive efficiency and to estimate the concentrations of EFA required by the lactating sow for maximum subsequent reproduction. Data were collected on 480 sows (PIC Camborough) balanced by parity, with 241 and 239 sows representing Parity 1, and 3-5 (P3+), respectively. Sows were assigned randomly, within parity, to a 3 × 3 factorial arrangement plus a control diet without added lipids. Factors included linoleic (2.1%, 2.7%, and 3.3%) and α-linolenic acid (0.15%, 0.30%, and 0.45%), obtained by adding 4% of different mixtures of canola, corn and flaxseed oils to diets. Diets were corn-soybean meal based with 12% wheat middlings. The benefits of supplemental EFA were more evident for the subsequent reproduction of mature P3+ sows. For these sows, supplemental α-linolenic acid improved the proportion of sows that farrowed relative to sows weaned (linear P=0.080; 82.8, 80.5, and 92.8% for sows fed 0.15%, 0.30%, and 0.45% α-linolenic acid, respectively). In addition, supplemental linoleic acid, fed to Parity 1 and P3+ sows, tended to increase subsequent litter size (linear P=0.074; 13.2, 13.8 and 14.0 total pigs born for 2.1%, 2.7% and 3.3% linoleic acid, respectively). These results demonstrate that a minimum dietary intake of both α-linolenic and linoleic acid is required for the modern lactating sow to achieve a maximum reproductive outcome through multiple mechanisms that include rapid return to estrus, increased maintenance of pregnancy and improved subsequent litter size.
Collapse
Affiliation(s)
- David S Rosero
- The Hanor Company, Franklin, KY 42134, USA; Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - R Dean Boyd
- The Hanor Company, Franklin, KY 42134, USA; Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | | | - Jack Odle
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Eric van Heugten
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA.
| |
Collapse
|
25
|
Wijendran V, Brenna JT, Wang DH, Zhu W, Meng D, Ganguli K, Kothapalli KSD, Requena P, Innis S, Walker WA. Long-chain polyunsaturated fatty acids attenuate the IL-1β-induced proinflammatory response in human fetal intestinal epithelial cells. Pediatr Res 2015; 78:626-33. [PMID: 26270575 PMCID: PMC5046822 DOI: 10.1038/pr.2015.154] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/13/2015] [Indexed: 11/09/2022]
Abstract
BACKGROUND Evidence suggests that excessive inflammation of the immature intestine may predispose premature infants to necrotizing enterocolitis (NEC). We investigated the anti-inflammatory effects of docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), and arachidonic acid (ARA) in human fetal and adult intestinal epithelial cells (IEC) in primary culture. METHODS Human fetal IEC in culture were derived from a healthy fetal small intestine (H4) or resected small intestine of a neonate with NEC (NEC-IEC). Intestinal cell lines Caco2 and NCM460 in culture were used as models for mature IEC. IEC in culture were pretreated with 100 µmol/l palmitic acid (PAL), DHA, EPA, ARA, or ARA+DHA for 48 h and then stimulated with proinflammatory IL-1β. RESULTS DHA significantly attenuated IL-1β induced proinflammatory IL-8 and IL-6 protein and mRNA in fetal H4, NEC-IEC, and mature Caco2, NCM460 IEC, compared to control and PAL treatment. DHA downregulated IL-1R1 (IL-1β receptor) and NFk β1 mRNA expression in fetal and adult IEC. ARA had potent anti-inflammatory effects with lower IL-8 and IL-6 (protein and mRNA) in fetal H4 but not in NEC-IEC or adult IEC. CONCLUSION The present study provides evidence that DHA and ARA may have important anti-inflammatory functions for prevention of NEC in premature infants.
Collapse
Affiliation(s)
- Vasuki Wijendran
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, USA
| | - JT Brenna
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Dong Hao Wang
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Weishu Zhu
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Di Meng
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Kriston Ganguli
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, USA
| | | | - Pilar Requena
- Liverpool School of Tropical Medicine, Liverpool, UK
| | - Sheila Innis
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - WA Walker
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, USA
| |
Collapse
|
26
|
Delarue J, Lallès JP. Nonalcoholic fatty liver disease: Roles of the gut and the liver and metabolic modulation by some dietary factors and especially long-chain n-3 PUFA. Mol Nutr Food Res 2015; 60:147-59. [DOI: 10.1002/mnfr.201500346] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 06/25/2015] [Accepted: 07/24/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Jacques Delarue
- Department of Nutritional Sciences; University Hospital and University of Brest; Brest France
- Breton Federation of Food and Human Nutrition (FED4216); University of Brest; Brest France
| | - Jean-Paul Lallès
- Breton Federation of Food and Human Nutrition (FED4216); University of Brest; Brest France
- Institut National de la Recherche Agronomique; UR1341; Alimentation et Adaptations Digestives; Nerveuses et Comportementales (ADNC); Saint-Gilles France
- Centre de Recherche en Nutrition Humaine-Ouest; Nantes Cedex 1 France
| |
Collapse
|
27
|
Liu Y. Fatty acids, inflammation and intestinal health in pigs. J Anim Sci Biotechnol 2015; 6:41. [PMID: 26361542 PMCID: PMC4564983 DOI: 10.1186/s40104-015-0040-1] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/28/2015] [Indexed: 12/13/2022] Open
Abstract
The intestine is not only critical for nutrient digestion and absorption, but also is the largest immune organ in the body. However, in pig production, inflammation induced by numerous factors, such as pathogen infection and stresses (e.g., weaning), results in intestinal mucosal injury and dysfunction, and consequently results in poor growth of pigs. Dietary fatty acids not only play critical roles in energy homeostasis and cellular membrane composition, but also exert potent effects on intestinal development, immune function, and inflammatory response. Recent studies support potential therapeutic roles for specific fatty acids (short chain and medium chain fatty acids and long chain polyunsaturated fatty acids) in intestinal inflammation of pigs. Results of these new lines of work indicate trophic and cytoprotective effects of fatty acids on intestinal integrity in pigs. In this article, we review the effect of inflammation on intestinal structure and function, and the role of specific fatty acids on intestinal health of pigs, especially under inflammatory conditions.
Collapse
Affiliation(s)
- Yulan Liu
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023 China
| |
Collapse
|
28
|
Porcine models of digestive disease: the future of large animal translational research. Transl Res 2015; 166:12-27. [PMID: 25655839 PMCID: PMC4458388 DOI: 10.1016/j.trsl.2015.01.004] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 01/03/2015] [Accepted: 01/07/2015] [Indexed: 12/14/2022]
Abstract
There is increasing interest in nonrodent translational models for the study of human disease. The pig, in particular, serves as a useful animal model for the study of pathophysiological conditions relevant to the human intestine. This review assesses currently used porcine models of gastrointestinal physiology and disease and provides a rationale for the use of these models for future translational studies. The pig has proven its utility for the study of fundamental disease conditions such as ischemia-reperfusion injury, stress-induced intestinal dysfunction, and short bowel syndrome. Pigs have also shown great promise for the study of intestinal barrier function, surgical tissue manipulation and intervention, as well as biomaterial implantation and tissue transplantation. Advantages of pig models highlighted by these studies include the physiological similarity to human intestine and mechanisms of human disease. Emerging future directions for porcine models of human disease include the fields of transgenics and stem cell biology, with exciting implications for regenerative medicine.
Collapse
|
29
|
Asparagine attenuates hepatic injury caused by lipopolysaccharide in weaned piglets associated with modulation of Toll-like receptor 4 and nucleotide-binding oligomerisation domain protein signalling and their negative regulators. Br J Nutr 2015; 114:189-201. [PMID: 26079268 DOI: 10.1017/s0007114515001476] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pro-inflammatory cytokines play a key role in many models of hepatic damage. In addition, asparagine (Asn) plays an important role in immune function. We aimed to investigate whether Asn could attenuate lipopolysaccharide (LPS)-induced liver damage. Forty-eight castrated barrows were allotted to four groups including: (1) non-challenged control; (2) LPS-challenged control; (3) LPS + 0.5% Asn; and (4) LPS + 1.0% Asn. After 19 d feeding with control, 0.5 or 1.0% Asn diets, pigs were injected with LPS or saline. Blood and liver samples were obtained at 4 h (early stage) and 24 h (late stage) post-injection. Asn alleviated liver injury, indicated by reduced serum aspartate aminotransferase and alkaline phosphatase activities linearly and quadratically; it increased claudin-1 protein expression linearly and quadratically at 24 h, and less severe liver morphological impairment at 4 or 24 h. In addition, Asn decreased mRNA expression of TNF-α and heat shock protein 70 (HSP70) linearly and quadratically at 4 h; it increased TNF-α mRNA expression, and HSP70 protein expression linearly and quadratically at 24 h. Moreover, Asn increased inducible NO synthase activity linearly and quadratically. Finally, Asn down-regulated the mRNA expression of Toll-like receptor 4 (TLR4) signalling molecules (TLR4, IL-1 receptor-associated kinase 1 (IRAK1), TNF-α receptor-associated factor 6), nucleotide-binding oligomerisation domain protein (NOD) signalling molecules (NOD1, NOD2 and their adaptor molecule receptor-interacting serine/threonine-protein kinase 2 (RIPK2)), and NF-κB p65 linearly or quadratically at 4 h. Oppositely, Asn up-regulated mRNA expressions of TLR4 and NOD signalling molecules (TLR4, myeloid differentiation factor 88, IRAK1, NOD2 and RIPK2), and their negative regulators (radioprotective 105, single Ig IL-1R-related molecule, Erbb2 interacting protein and centaurin β1) linearly or quadratically at 24 h. These results indicate that, in early and late stages of LPS challenge, Asn improves liver integrity and exerts different regulatory effects on mRNA expression of TLR4 and NOD signalling molecules.
Collapse
|
30
|
Rosero DS, Odle J, Mendoza SM, Boyd RD, Fellner V, van Heugten E. Impact of dietary lipids on sow milk composition and balance of essential fatty acids during lactation in prolific sows. J Anim Sci 2015; 93:2935-47. [PMID: 26115280 DOI: 10.2527/jas.2014-8529] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
Two studies were designed to determine the effects of supplementing diets with lipid sources of EFA (linoleic and α-linolenic acid) on sow milk composition to estimate the balance of EFA for sows nursing large litters. In Exp. 1, 30 sows, equally balanced by parity (1 and 3 to 5) and nursing 12 pigs, were fed diets supplemented with 6% animal-vegetable blend (A-V), 6% choice white grease (CWG), or a control diet without added lipid. Diets were corn-soybean meal based with 8% corn distiller dried grains with solubles and 6% wheat middlings and contained 3.25 g standardized ileal digestible Lys/Mcal ME. Sows fed lipid-supplemented diets secreted greater amounts of fat (P = 0.082; 499 and 559 g/d for control and lipid-added diets, respectively) than sows fed the control diet. The balance of EFA was computed as apparent ileal digestible intake of EFA minus the outflow of EFA in milk. For sows fed the control diet, the amount of linoleic acid secreted in milk was greater than the amount consumed, throughout lactation. This resulted in a pronounced negative balance of linoleic acid (-22.4, -38.0, and -14.1 g/d for d 3, 10, and 17 of lactation, respectively). In Exp. 2, 50 sows, equally balanced by parity and nursing 12 pigs, were randomly assigned to a 2 × 2 factorial arrangement of diets plus a control diet without added lipids. Factors included linoleic acid (2.1% and 3.3%) and α-linolenic acid (0.15% and 0.45%). The different concentrations of EFA were obtained by adding 4% of different mixtures of canola, corn, and flaxseed oils to diets. The n-6 to n-3 fatty acid ratios in the diets ranged from 5 to 22. Increasing supplemental EFA increased (P < 0.001) milk concentrations of linoleic (16.7% and 20.8%, for 2.1% and 3.3% linoleic acid, respectively) and α-linolenic acid (P < 0.001; 1.1 and 1.9% for 0.15 and 0.45% α-linolenic acid, respectively). Increasing supplemental EFA increased the estimated balance of α-linolenic acid (P < 0.001; -0.2 and 5.3 g/d for 0.15% and 0.45% α-linolenic acid, respectively), but not linoleic acid (P = 0.14; -3.4 and 10.0 g/d for 2.1% and 3.3% linoleic acid, respectively). In conclusion, lipid supplementation to sow lactation diets improved milk fat secretion. The fatty acid composition of milk fat reflected the dietary supplementation of EFA. The net effect of supplemental EFA was to create a positive balance during lactation, which may prove to be beneficial for the development of nursing piglets and the subsequent reproduction of sows.
Collapse
|
31
|
Oh SY, Lee SJ, Jung YH, Lee HJ, Han HJ. Arachidonic acid promotes skin wound healing through induction of human MSC migration by MT3-MMP-mediated fibronectin degradation. Cell Death Dis 2015; 6:e1750. [PMID: 25950480 PMCID: PMC4669694 DOI: 10.1038/cddis.2015.114] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/09/2015] [Accepted: 03/23/2015] [Indexed: 12/17/2022]
Abstract
Arachidonic acid (AA) is largely released during injury, but it has not been fully studied yet how AA modulates wound repair with stem cells. Therefore, we investigated skin wound-healing effect of AA-stimulated human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) in vivo and its molecular mechanism in vitro. We found that transplantation of hUCB-MSCs pre-treated with AA enhanced wound filling, re-epithelization, and angiogenesis in a mouse skin excisional wound model. AA significantly promoted hUCB-MSCs migration after a 24 h incubation, which was inhibited by the knockdown of G-protein-coupled receptor 40 (GPR40). AA activated mammalian target of rapamycin complex 2 (mTORC2) and Aktser473 through the GPR40/phosphoinositide 3-kinase (PI3K) signaling, which was responsible for the stimulation of an atypical protein kinase C (PKC) isoform, PKCζ. Subsequently, AA stimulated phosphorylation of p38 MAPK and transcription factor Sp1, and induced membrane type 3-matrix metalloproteinase (MT3-MMP)-dependent fibronectin degradation in promoting hUCB-MSCs motility. Finally, the silencing of MT3-MMP in AA-stimulated hUCB-MSCs failed to promote the repair of skin wounds owing to impaired cell motility. In conclusion, AA enhances skin wound healing through induction of hUCB-MSCs motility by MT3-MMP-mediated fibronectin degradation, which relies on GPR40-dependent mTORC2 signaling pathways.
Collapse
Affiliation(s)
- S Y Oh
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, 151-741, Korea
| | - S-J Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, 151-741, Korea
| | - Y H Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, 151-741, Korea
| | - H J Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, 151-741, Korea
| | - H J Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, 151-741, Korea
| |
Collapse
|
32
|
Gonzalez LM, Moeser AJ, Blikslager AT. Animal models of ischemia-reperfusion-induced intestinal injury: progress and promise for translational research. Am J Physiol Gastrointest Liver Physiol 2015; 308:G63-75. [PMID: 25414098 PMCID: PMC4297854 DOI: 10.1152/ajpgi.00112.2013] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Research in the field of ischemia-reperfusion injury continues to be plagued by the inability to translate research findings to clinically useful therapies. This may in part relate to the complexity of disease processes that result in intestinal ischemia but may also result from inappropriate research model selection. Research animal models have been integral to the study of ischemia-reperfusion-induced intestinal injury. However, the clinical conditions that compromise intestinal blood flow in clinical patients ranges widely from primary intestinal disease to processes secondary to distant organ failure and generalized systemic disease. Thus models that closely resemble human pathology in clinical conditions as disparate as volvulus, shock, and necrotizing enterocolitis are likely to give the greatest opportunity to understand mechanisms of ischemia that may ultimately translate to patient care. Furthermore, conditions that result in varying levels of ischemia may be further complicated by the reperfusion of blood to tissues that, in some cases, further exacerbates injury. This review assesses animal models of ischemia-reperfusion injury as well as the knowledge that has been derived from each to aid selection of appropriate research models. In addition, a discussion of the future of intestinal ischemia-reperfusion research is provided to place some context on the areas likely to provide the greatest benefit from continued research of ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Liara M. Gonzalez
- 1Department of Clinical Sciences, Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina; and
| | - Adam J. Moeser
- 2Department of Population Health and Pathobiology, Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina
| | - Anthony T. Blikslager
- 1Department of Clinical Sciences, Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina; and
| |
Collapse
|
33
|
Abstract
Advanced mucosal healing (MH) after intestinal mucosal inflammation coincides with sustained clinical remission and reduced rates of hospitalization and surgical resection, explaining why MH is increasingly considered as a full therapeutic goal and as an endpoint for clinical trials. Intestinal MH is a complex phenomenon viewed as a succession of steps necessary to restore tissue structure and function. These steps include epithelial cell migration and proliferation, cell differentiation, restoration of epithelial barrier functions, and modulation of cell apoptosis. Few clinical studies have evaluated the needs for specific macronutrients and micronutrients and their effects on intestinal MH, most data having been obtained from animal and cell studies. These data suggest that supplementation with specific amino acids including arginine, glutamine, glutamate, threonine, methionine, serine, proline, and the amino acid-derived compounds, polyamines can favorably influence MH. Short-chain fatty acids, which are produced by the microbiota from undigested polysaccharides and protein-derived amino acids, also exert beneficial effects on the process of intestinal MH in experimental models. Regarding supplementation with lipids, although the effects of ω-3 and ω-6 fatty acids remain controversial, endogenous prostaglandin synthesis seems to be necessary for MH. Finally, among micronutrients, several vitamin and mineral deficiencies with different frequencies have been observed in patients with inflammatory bowel diseases and supplementation with some of them (vitamin A, vitamin D3, vitamin C, and zinc) are presumed to favor MH. Future work, including clinical studies, should evaluate the efficiency of supplementation with combination of dietary compounds as adjuvant nutritional intervention for MH of the inflamed intestinal mucosa.
Collapse
|
34
|
Feng Y, Browner P, Teitelbaum DH. Effects on varying intravenous lipid emulsions on the small bowel epithelium in a mouse model of parenteral nutrition. JPEN J Parenter Enteral Nutr 2013; 37:775-86. [PMID: 23757306 DOI: 10.1177/0148607113491608] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Injectable fat emulsions (FEs) are a clinically dependable source of essential fatty acids (FA). ω-6 FA is associated with an inflammatory response. Medium-chain triglycerides (MCT, ω-3 FA), fish oil, and olive oil are reported to decrease the inflammatory response. However, the effect of these lipids on the gastrointestinal tract has not been well studied. To address this, we used a mouse model of parenteral nutrition (PN) and hypothesized that a decrease in intestinal inflammation would be seen when either fish oil and MCT or olive oil were added. METHODS Three FEs were studied in adult C57BL/6 mice via intravenous cannulation: standard soybean-based FE (SBFE), 80% olive oil -supplemented FE (OOFE), or a combination of a soybean oil, MCT, olive oil, and fish oil emulsion (SMOF). PN was given for 7 days, small bowel mucosa-derived cytokines, animal survival rate, epithelial cell (EC) proliferation and apoptosis rates, intestinal barrier function and mucosal FA composition were analyzed. RESULTS Compared to the SBFE and SMOF groups, the best survival, highest EC proliferation and lowest EC apoptosis rates were observed in the OOFE group; and associated with the lowest levels of tumor necrosis factor-α, interleukin-6, and interleukin-1β expression. Jejunal FA content showed higher levels of eicosapentaenoic and docosapentaenoic acid in the SMOF group and the highest arachidonic acid in the OOFE group. CONCLUSION The study showed that PN containing OOFE had beneficial effects to small bowel health and animal survival. Further investigation may help to enhance bowel integrity in patients restricted to PN.
Collapse
Affiliation(s)
- Yongjia Feng
- Section of Pediatric Surgery, Department of Surgery, the University of Michigan Medical School and the C. S. Mott Children's Hospital, Ann Arbor, Michigan
| | | | | |
Collapse
|
35
|
The growing role of eicosanoids in tissue regeneration, repair, and wound healing. Prostaglandins Other Lipid Mediat 2013; 104-105:130-8. [PMID: 23727457 DOI: 10.1016/j.prostaglandins.2013.05.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Revised: 04/14/2013] [Accepted: 05/08/2013] [Indexed: 11/21/2022]
Abstract
Tissue repair and regeneration are essential processes in maintaining tissue homeostasis, especially in response to injury or stress. Eicosanoids are ubiquitous mediators of cell proliferation, differentiation, and angiogenesis, all of which are important for tissue growth. Eicosanoids regulate the induction and resolution of inflammation that accompany the tissue response to injury. In this review, we describe how this diverse group of molecules is a key regulator of tissue repair and regeneration in multiple organ systems and biologic contexts.
Collapse
|
36
|
Chen F, Liu Y, Zhu H, Hong Y, Wu Z, Hou Y, Li Q, Ding B, Yi D, Chen H. Fish oil attenuates liver injury caused by LPS in weaned pigs associated with inhibition of TLR4 and nucleotide-binding oligomerization domain protein signaling pathways. Innate Immun 2013; 19:504-15. [DOI: 10.1177/1753425912472003] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
This study evaluated whether fish oil exerted a hepatoprotective effect in a LPS-induced liver injury model via regulation of TLR4 and nucleotide-binding oligomerization domain protein (NOD) signaling pathways. Twenty-four piglets were used in a 2 × 2 factorial design, and the main factors included diet (5% corn oil or 5% fish oil) and immunological challenge (LPS or saline). Fish oil resulted in enrichment of eicosapentaenoic acid, docosahexaenoic acid and total (n-3) polyunsaturated fatty acids in liver. Less severe liver injury was observed in pigs fed fish oil, as evidenced by improved serum biochemical parameters and less severe histological liver damage. In addition, higher expression of liver tight junction proteins, and lower hepatocyte proliferation and higher hepatocyte apoptosis were observed in pigs fed fish oil. The improved liver integrity in pigs fed fish oil was concurrent with reduced hepatic mRNA expression of TLR4, myeloid differentiation factor 88, IL-1 receptor-associated kinase 1 and TNF- α receptor-associated factor 6, and NOD1, NOD2 and receptor-interacting serine/threonine-protein kinase 2, as well as reduced hepatic protein expression of NF-κB p65, leading to reduced hepatic pro-inflammatory mediators. These results indicate that fish oil improves liver integrity partially via inhibition of TLR4 and NOD signaling pathways under an inflammatory condition.
Collapse
Affiliation(s)
- Feng Chen
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People’s Republic of China
| | - Yulan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People’s Republic of China
| | - Huiling Zhu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People’s Republic of China
| | - Yu Hong
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People’s Republic of China
| | - Zhifeng Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People’s Republic of China
| | - Yongqing Hou
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People’s Republic of China
| | - Quan Li
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People’s Republic of China
| | - Binying Ding
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People’s Republic of China
| | - Dan Yi
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People’s Republic of China
| | - Hongbo Chen
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People’s Republic of China
| |
Collapse
|
37
|
Liu Y, Chen F, Odle J, Lin X, Jacobi SK, Zhu H, Wu Z, Hou Y. Fish oil enhances intestinal integrity and inhibits TLR4 and NOD2 signaling pathways in weaned pigs after LPS challenge. J Nutr 2012; 142:2017-24. [PMID: 23014495 DOI: 10.3945/jn.112.164947] [Citation(s) in RCA: 211] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Long-chain (n-3) PUFA exert beneficial effects on inflammatory bowel diseases in animal models and clinical trials. In addition, pattern recognition receptors such as toll-like receptors (TLR) and nucleotide-binding oligomerization domain proteins (NOD) play a critical role in intestinal inflammation. We hypothesized that fish oil could alleviate Escherichia coli LPS-induced intestinal injury via modulation of TLR4 and NOD signaling pathways. Twenty-four weaned piglets were used in a 2 × 2 factorial design and the main factors included a dietary treatment (5% corn oil or 5% fish oil) and immunological challenge (LPS or saline). After feeding fish oil or corn oil diets for 21 d, pigs were injected with LPS or saline. At 4 h postinjection, blood samples were collected and pigs were killed. EPA, DHA, and total (n-3) PUFA were enriched in intestinal mucosa through fish supplementation. Fish oil improved intestinal morphology, indicated by greater villus height and villus height:crypt depth ratio, and intestinal barrier function, indicated by decreased plasma diamine oxidase (DAO) activity and increased mucosal DAO activity as well as enhanced protein expression of intestinal tight junction proteins including occludin and claudin-1. Moreover, fish oil decreased intestinal TNFα and PGE(2) concentrations and caspase-3 and heat shock protein 70 protein expression. Finally, fish oil downregulated the mRNA expression of intestinal TLR4 and its downstream signals myeloid differentiation factor 88, IL-1 receptor-associated kinase 1, TNFα receptor-associated factor 6, and NOD2, and its adaptor molecule, receptor-interacting serine/threonine-protein kinase 2. Fish oil decreased the protein expression of intestinal NFκB p65. These results indicate that fish oil supplementation is associated with inhibition of TLR4 and NOD2 signaling pathways and concomitant improvement of intestinal integrity under an inflammatory condition.
Collapse
Affiliation(s)
- Yulan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Dietary nutrients are essential for gastrointestinal (GI) growth and function, and nutritional support of GI growth and development is a significant component of infant care. For healthy full-term neonates, nutritional provisions of the mother's milk and/or formula will support normal maturation of structure and function of the GI tract in most infants. The composition of breast milk affects GI barrier function and development of a competent mucosal immune system. The functional nutrients and other bioactive components of milk support a microenvironment for gut protection and maturation. However, premature infants struggle with feeding tolerance impairing normal GI function, leading to intestinal dysfunction and even death. The high prevalence worldwide of enteric diseases and dysfunction in neonates has led to much interest in understanding the role of nutrients and food components in the establishment and maintenance of a functioning GI tract. Neonates who do not receive enteral feeding as either mother's milk or formula are supported by total parental nutrition (TPN). The lack of enteral nutrition can compound intestinal dysfunction, leading to high morbidity and mortality in intestinally compromised infants. Reciprocally, enteral stimulation of an immature GI tract can also compound intestinal dysfunction. Therefore, further understanding of nutrient interactions with the mucosa is necessary to define nutritional requirements of the developing GI tract to minimize intestinal complications and infant morbidity. Piglet models of intestinal development and function are similar to humans, and this review summarizes recent findings regarding nutrient requirements for growth and maintenance of intestinal health. In particular, this article reviews the role of specific amino acids (arginine, glutamine, glutamate, and threonine), fatty acids (long chain polyunsaturated, medium chain, and short chain), various prebiotic carbohydrates (short-chain fructo-oligosaccharide, fructo--oligosaccharide, lacto-N-neotetraose, human milk oligosaccharide, polydextrose, and galacto-oligosaccharide), and probiotics that have been examined in the suckling piglet model of intestinal health.
Collapse
Affiliation(s)
- Sheila K Jacobi
- Laboratory of Developmental Nutrition, Department of Animal Science, North Carolina State University, Raleigh, NC, USA
| | | |
Collapse
|