1
|
Drenckpohl DC, Christifano DN, Carlson SE. Is choline deficiency an unrecognized factor in necrotizing enterocolitis of preterm infants? Pediatr Res 2024; 96:875-883. [PMID: 38658665 DOI: 10.1038/s41390-024-03212-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/23/2024] [Accepted: 03/15/2024] [Indexed: 04/26/2024]
Abstract
We undertook this review to determine if it is plausible that choline or phosphatidylcholine (PC) deficiency is a factor in necrotizing enterocolitis (NEC) after two clinical trials found a dramatic and unexpected reduction in NEC in an experimental group provided higher PC compared to a control group. Sources and amounts of choline/PC for preterm infants are compared to the choline status of preterm infants at birth and following conventional nutritional management. The roles of choline/PC in intestinal structure, mucus, mesenteric blood flow, and the cholinergic anti-inflammatory system are summarized. Low choline/PC status is linked to prematurity/immaturity, parenteral and enteral feeding, microbial dysbiosis and hypoxia/ischemia, factors long associated with the risk of developing NEC. We conclude that low choline status exists in preterm infants provided conventional parenteral and enteral nutritional management, and that it is plausible low choline/PC status adversely affects intestinal function to set up the vicious cycle of inflammation, loss of intestinal barrier function and worsening tissue hypoxia that occurs with NEC. In conclusion, this review supports the need for randomized clinical trials to test the hypothesis that additional choline or PC provided parenterally or enterally can reduce the incidence of NEC in preterm infants. IMPACT STATEMENT: Low choline status in preterm infants who are managed by conventional nutrition is plausibly linked to the risk of developing necrotizing enterocolitis.
Collapse
Affiliation(s)
- Douglas C Drenckpohl
- Department of Food & Nutrition, OSF Healthcare Saint Francis Medical Center, Peoria, IL, 61637, USA
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, 66106, USA
| | - Danielle N Christifano
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, 66106, USA
| | - Susan E Carlson
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, 66106, USA.
| |
Collapse
|
2
|
Gershner GH, Hunter CJ. Redox Chemistry: Implications for Necrotizing Enterocolitis. Int J Mol Sci 2024; 25:8416. [PMID: 39125983 PMCID: PMC11312856 DOI: 10.3390/ijms25158416] [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: 06/18/2024] [Revised: 07/22/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Reduction-oxidation (redox) chemistry plays a vital role in human homeostasis. These reactions play critical roles in energy generation, as part of innate immunity, and in the generation of secondary messengers with various functions such as cell cycle progression or the release of neurotransmitters. Despite this cornerstone role, if left unchecked, the body can overproduce reactive oxygen species (ROS) or reactive nitrogen species (RNS). When these overwhelm endogenous antioxidant systems, oxidative stress (OS) occurs. In neonates, OS has been associated with retinopathy of prematurity (ROP), leukomalacia, and bronchopulmonary dysplasia (BPD). Given its broad spectrum of effects, research has started to examine whether OS plays a role in necrotizing enterocolitis (NEC). In this paper, we will discuss the basics of redox chemistry and how the human body keeps these in check. We will then discuss what happens when these go awry, focusing mostly on NEC in neonates.
Collapse
Affiliation(s)
- Grant H. Gershner
- Division of Pediatric Surgery, Oklahoma Children’s Hospital, 1200 Everett Drive, ET NP 2320, Oklahoma City, OK 73104, USA;
- Department of Surgery, The University of Oklahoma Health Sciences Center, 800 Research Parkway, Suite 449, Oklahoma City, OK 73104, USA
| | - Catherine J. Hunter
- Division of Pediatric Surgery, Oklahoma Children’s Hospital, 1200 Everett Drive, ET NP 2320, Oklahoma City, OK 73104, USA;
- Department of Surgery, The University of Oklahoma Health Sciences Center, 800 Research Parkway, Suite 449, Oklahoma City, OK 73104, USA
| |
Collapse
|
3
|
Zhong H, Xie Q, Li F, Yang Z, Li K, Luo Q. Determination of oxylipins and their precursors in breast milk by solid phase extraction-ultra high performance liquid chromatography-triple quadrupole tandem mass spectrometry. J Chromatogr A 2023; 1709:464400. [PMID: 37769518 DOI: 10.1016/j.chroma.2023.464400] [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: 07/16/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/03/2023]
Abstract
Oxylipins and their precursors (long-chain polyunsaturated fatty acids, LCPUFAs) are key intercellular signaling molecules influencing the inflammatory response. Each oxylipin has pro- and/or anti-inflammatory effects, and the relative abundance of different oxylipins can alter the inflammatory balance, making it important to clarify the oxylipin profile of breast milk for optimal infant health. The extraction, identification, and simultaneous quantification of oxylipins in breast milk are challenging due to the structural similarity, limited stability, and the low endogenous concentration of oxylipins and the complex matrix of breast milk. This study aimed to develop a solid phase extraction-ultra high performance liquid chromatography-triple quadrupole tandem mass spectrometry (SPE-UPLC-MS/MS) method for the comprehensive and specific quantification of oxylipins and their precursors in breast milk. The LC conditions (including column, mobile phase, and gradient conditions) and SPE procedure (including SPE cartridges, elution solvent, and elution volume) were optimized to achieve accurate quantification and better analyte recovery. A single 18-minute chromatographic run allows for the quantification of 20 oxylipins and 5 PUFAs. The results showed good linearity (R2 > 0.99) over the concentration range of 2 to 100 ng/mL, with the instrument detection limits ranging from 0.01 to 0.90 ng/mL for oxylipins and 0.02 to 0.59 ng/mL for PUFAs. The method is rapid, sensitive, and reproducible (RSD ≤ 10%) and is suitable for the quantitative analysis of oxylipins and their precursors in infant formula samples.
Collapse
Affiliation(s)
- Huifang Zhong
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Qinggang Xie
- Heilongjiang Feihe Dairy Co., Ltd., Qiqihaer 164800, China
| | - Fang Li
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhiyi Yang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Kaifeng Li
- Heilongjiang Feihe Dairy Co., Ltd., Qiqihaer 164800, China
| | - Qian Luo
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
4
|
Seethaler B, Lehnert K, Yahiaoui-Doktor M, Basrai M, Vetter W, Kiechle M, Bischoff SC. Omega-3 polyunsaturated fatty acids improve intestinal barrier integrity-albeit to a lesser degree than short-chain fatty acids: an exploratory analysis of the randomized controlled LIBRE trial. Eur J Nutr 2023; 62:2779-2791. [PMID: 37318580 PMCID: PMC10468946 DOI: 10.1007/s00394-023-03172-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 05/04/2023] [Indexed: 06/16/2023]
Abstract
PURPOSE Adherence to the Mediterranean diet is associated with beneficial health effects, including gastrointestinal disorders. Preclinical studies suggest that omega-3 polyunsaturated fatty acids (n-3 PUFAs), found in Mediterranean foods like nuts and fish, improve intestinal barrier integrity. Here, we assessed possible effects of n-3 PUFAs on barrier integrity in a randomized controlled trial. METHODS We studied 68 women from the open-label LIBRE trial (clinicaltrials.gov: NCT02087592) who followed either a Mediterranean diet (intervention group, IG) or a standard diet (control group, CG). Study visits comprised baseline, month 3, and month 12. Barrier integrity was assessed by plasma lipopolysaccharide binding protein (LBP) and fecal zonulin; fatty acids by gas chromatography with mass spectrometry. Median and interquartile ranges are shown. RESULTS Adherence to the Mediterranean diet increased the proportion of the n-3 docosahexaenoic acid (DHA) (IG + 1.5% [0.9;2.5, p < 0.001]/ + 0.3% [- 0.1;0.9, p < 0.050] after 3/12 months; CG + 0.9% [0.5;1.6, p < 0.001]/ ± 0%) and decreased plasma LBP (IG - 0.3 µg/ml [- 0.6;0.1, p < 0.010]/ - 0.3 µg/ml [- 1.1; - 0.1, p < 0.001]; CG - 0.2 µg/ml [- 0.8; - 0.1, p < 0.001]/ ± 0 µg/ml) and fecal zonulin levels (IG - 76 ng/mg [- 164; - 12, p < 0.010]/ - 74 ng/mg [- 197;15, p < 0.001]; CG - 59 ng/mg [- 186;15, p < 0.050]/ + 10 ng/mg [- 117;24, p > 0.050]). Plasma DHA and LBP (R2: 0.14-0.42; all p < 0.070), as well as plasma DHA and fecal zonulin (R2: 0.18-0.48; all p < 0.050) were found to be inversely associated in bi- and multivariate analyses. Further multivariate analyses showed that the effect of DHA on barrier integrity was less pronounced than the effect of fecal short-chain fatty acids on barrier integrity. CONCLUSIONS Our data show that n-3 PUFAs can improve intestinal barrier integrity. TRIAL REGISTRATION NUMBER The trial was registered prospectively at ClinicalTrials.gov (reference: NCT02087592).
Collapse
Affiliation(s)
- Benjamin Seethaler
- Institute of Nutritional Medicine, University of Hohenheim, Fruwirthstr. 12, 70593, Stuttgart, Germany
| | - Katja Lehnert
- Institute of Food Chemistry, University of Hohenheim, Stuttgart, Germany
| | - Maryam Yahiaoui-Doktor
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany
| | - Maryam Basrai
- Institute of Nutritional Medicine, University of Hohenheim, Fruwirthstr. 12, 70593, Stuttgart, Germany
| | - Walter Vetter
- Institute of Food Chemistry, University of Hohenheim, Stuttgart, Germany
| | - Marion Kiechle
- Department of Gynecology, Center for Hereditary Breast and Ovarian Cancer, Klinikum Rechts der Isar, Technical University Munich and Comprehensive Cancer Center Munich, Munich, Germany
| | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Fruwirthstr. 12, 70593, Stuttgart, Germany.
| |
Collapse
|
5
|
Das UN. Infection, Inflammation, and Immunity in Sepsis. Biomolecules 2023; 13:1332. [PMID: 37759732 PMCID: PMC10526286 DOI: 10.3390/biom13091332] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Sepsis is triggered by microbial infection, injury, or even major surgery. Both innate and adaptive immune systems are involved in its pathogenesis. Cytoplasmic presence of DNA or RNA of the invading organisms or damaged nuclear material (in the form of micronucleus in the cytoplasm) in the host cell need to be eliminated by various nucleases; failure to do so leads to the triggering of inflammation by the cellular cGAS-STING system, which induces the release of IL-6, TNF-α, and IFNs. These cytokines activate phospholipase A2 (PLA2), leading to the release of polyunsaturated fatty acids (PUFAs), gamma-linolenic acid (GLA), arachidonic acid (AA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), which form precursors to various pro- and anti-inflammatory eicosanoids. On the other hand, corticosteroids inhibit PLA2 activity and, thus, suppress the release of GLA, AA, EPA, and DHA. PUFAs and their metabolites have a negative regulatory action on the cGAS-STING pathway and, thus, suppress the inflammatory process and initiate inflammation resolution. Pro-inflammatory cytokines and corticosteroids (corticosteroids > IL-6, TNF-α) suppress desaturases, which results in decreased formation of GLA, AA, and other PUFAs from the dietary essential fatty acids (EFAs). A deficiency of GLA, AA, EPA, and DHA results in decreased production of anti-inflammatory eicosanoids and failure to suppress the cGAS-STING system. This results in the continuation of the inflammatory process. Thus, altered concentrations of PUFAs and their metabolites, and failure to suppress the cGAS-STING system at an appropriate time, leads to the onset of sepsis. Similar abnormalities are also seen in radiation-induced inflammation. These results imply that timely administration of GLA, AA, EPA, and DHA, in combination with corticosteroids and anti-IL-6 and anti-TNF-α antibodies, may be of benefit in mitigating radiation-induced damage and sepsis.
Collapse
Affiliation(s)
- Undurti N Das
- UND Life Sciences, 2221 NW 5th St., Battle Ground, WA 98604, USA
| |
Collapse
|
6
|
Sami AS, Frazer LC, Miller CM, Singh DK, Clodfelter LG, Orgel KA, Good M. The role of human milk nutrients in preventing necrotizing enterocolitis. Front Pediatr 2023; 11:1188050. [PMID: 37334221 PMCID: PMC10272619 DOI: 10.3389/fped.2023.1188050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is an intestinal disease that primarily impacts preterm infants. The pathophysiology of NEC involves a complex interplay of factors that result in a deleterious immune response, injury to the intestinal mucosa, and in its most severe form, irreversible intestinal necrosis. Treatments for NEC remain limited, but one of the most effective preventative strategies for NEC is the provision of breast milk feeds. In this review, we discuss mechanisms by which bioactive nutrients in breast milk impact neonatal intestinal physiology and the development of NEC. We also review experimental models of NEC that have been used to study the role of breast milk components in disease pathophysiology. These models are necessary to accelerate mechanistic research and improve outcomes for neonates with NEC.
Collapse
Affiliation(s)
- Ahmad S. Sami
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lauren C. Frazer
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Claire M. Miller
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Dhirendra K. Singh
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lynda G. Clodfelter
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kelly A. Orgel
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Misty Good
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
7
|
Alshaikh BN, Reyes Loredo A, Yusuf K, Maarouf A, Fenton TR, Momin S. Enteral long-chain polyunsaturated fatty acids and necrotizing enterocolitis: A systematic review and meta-analysis. Am J Clin Nutr 2023; 117:918-929. [PMID: 37137615 DOI: 10.1016/j.ajcnut.2023.01.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND Preterm infants are at risk of long-chain polyunsaturated fatty acid (LCPUFA) deficiency. Recent studies on high-dose DHA; n-3 LCPUFA in preterm infants suggested potential positive effects on cognitive outcomes but raised concerns about some increased neonatal morbidities. These studies and recent recommendations for DHA supplementation generated controversy owing to the lack of balance between DHA and arachidonic acid (ARA; n-6 LCPUFA). OBJECTIVES To identify the effect of enteral supplementation of DHA, with and without ARA, on necrotizing enterocolitis (NEC) in very preterm infants. METHODS A systematic review of randomized and controlled trials compared enteral LCPUFAs with placebo or no supplementation in very preterm infants. We searched PubMed, Ovid-MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials, and CINHAL databases from inception to July 2022. Data were extracted in duplicate using a structured proforma. A meta-analysis and metaregression with random-effects models were used. The interventions evaluated were DHA alone vs. that combined with ARA, source of DHA, dose, and supplement delivery methods. Methodological qualities and risk of bias were assessed using the Cochrane risk-of-bias tool. RESULTS Fifteen randomized clinical trials (RCTs) included 3963 very preterm infants with 217 cases of NEC. Supplementation with DHA alone increased NEC (2620 infants; RR: 1.56; 95% CI: 1.02, 2.39) with no evidence of heterogeneity (I2 = 0.0%, P = 0.46). Multiple metaregression revealed significant reduction in NEC when ARA was supplemented with DHA (aRR 0.42; 95% CI: 0.21, 0.88). The source of DHA, dose, and feeding type revealed no associations with NEC. Two RCTs supplemented high-dose DHA to lactating mothers. There was a significant increase in risk of NEC with this approach (1148 infants; RR: 1.92; 95% CI: 1.02, 3.61) with no evidence of heterogeneity (I2 = 0.0, P = 0.81). CONCLUSIONS Supplementation with DHA alone may increase risk of NEC. Concurrent supplementation with ARA needs to be considered when adding DHA to preterm infants' diet.
Collapse
Affiliation(s)
- Belal N Alshaikh
- Neonatal Nutrition and Gastroenterology Program, Cumming School of Medicine, University of Calgary, Calgary AB, Canada; Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary AB, Canada.
| | - Adriana Reyes Loredo
- Neonatal Nutrition and Gastroenterology Program, Cumming School of Medicine, University of Calgary, Calgary AB, Canada; Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| | - Kamran Yusuf
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| | - Ahmed Maarouf
- Neonatal Nutrition and Gastroenterology Program, Cumming School of Medicine, University of Calgary, Calgary AB, Canada; Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| | - Tanis R Fenton
- Community Health Sciences, Institute of Public Health, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary AB, Canada; Nutrition Services, Alberta Health Services, Calgary AB, Canada
| | - Sarfaraz Momin
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| |
Collapse
|
8
|
Liu Y, Hoang TK, Park ES, Freeborn J, Okeugo B, Tran DQ, Rhoads JM. Probiotic-educated Tregs are more potent than naïve Tregs for immune tolerance in stressed new-born mice. Benef Microbes 2023; 14:73-84. [PMID: 36815493 PMCID: PMC10124588 DOI: 10.3920/bm2022.0095] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
When new-born mice are subjected to acute maternal separation stress, cow-milk based formula feeding, and brief recurrent hypoxia with cold stress, they develop gut inflammation similar to the phenotype of neonatal necrotizing enterocolitis, characterised by an increase in gut mucosal effector T (Teffs) and reduced Foxp3+ regulatory T (Tregs) cells. The imbalance can be prevented by probiotic Limosilactobacillus reuteri DSM 17938 (LR 17938). We hypothesised that LR 17938 could potentiate a tolerogenic function of Tregs. To analyse whether LR 17938 can educate Tregs to improve their tolerogenic potency during neonatal stress, we isolated T cells (Tregs and Teffs) from 'donor' mice fed with either LR 17938 (107 cfu) or control media. The cells were adoptively transferred (AT) by intraperitoneal injection (5 × 105 cells/mouse) to new-born (d5) recipient mice. Mice were then separated from their dams, fed formula by gavage, and exposed to hypoxia and cold stress (NeoStress) for 4 days. We analysed the percentage of Tregs in CD4+T helper cells in the intestine (INT) and mesenteric lymph nodes (MLN) of recipient mice. We found that: (1) the percentage of Tregs in the INT and MLN following NeoStress were significantly reduced compared to dam-fed unstressed mice; (2) AT of either naïve Tregs or LR-educated Tregs to mice with Neostress increased the percentage of Tregs in the INT and MLN compared to the percentage in NeoStress mice without Treg treatment; however, LR-educated Tregs increased the Tregs significantly more than naïve Tregs; and (3) AT of LR-educated Tregs reduced pro-inflammatory CD44+Foxp3-NonTregs and inflammatory CX3CR1+ dendritic cells in the intestinal mucosa of NeoStress mice. In conclusion, adoptive transfer of Tregs promotes the generation of and/or migration of endogenous Tregs in the intestinal mucosa of recipient mice. Importantly, probiotic-educated Tregs are more potent than naïve Tregs to enhance immune tolerance following neonatal stress.
Collapse
Affiliation(s)
- Y Liu
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - T K Hoang
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - E S Park
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - J Freeborn
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - B Okeugo
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - D Q Tran
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - J M Rhoads
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| |
Collapse
|
9
|
Welboren AC, Van Baal J, Ghaffari MH, Sauerwein H, Renaud JB, Martín-Tereso J, Steele MA, Leal LN. Gastrointestinal permeability and inflammatory status of preweaning dairy calves in response to decreasing the ratio of n-6 to n-3 fatty acid in milk replacer. J Dairy Sci 2023; 106:3662-3679. [PMID: 37002139 DOI: 10.3168/jds.2022-22568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 12/05/2022] [Indexed: 03/31/2023]
Abstract
The ratio of n-6 to n-3 fatty acid (FA) is between 2 and 10 times higher in milk replacer (MR) than in whole milk, which may promote inflammation and compromise the integrity of the intestinal epithelium. To evaluate how decreasing the n-6:n-3 FA ratio of MR affects gastrointestinal (GIT) permeability and inflammatory status, 30 dairy calves (2.8 ± 1.06 d of age; mean ± standard deviation) were randomly assigned to be fed an MR with an n-6:n-3 FA ratio of 40:1 (CON; 29.3% crude fat of DM; n = 15) or 6.5:1 (n-3; 29.1% crude fat of DM; n = 15). Calves were fed 7.0 L/d in 2 meals. Calves were weighed and fecal consistency was analyzed weekly. On d 22, calves were administered Cr-EDTA, lactulose, and d-mannitol to assess GIT permeability. Blood and total urine were sequentially collected for 6 and 24 h, respectively, and analyzed for marker content. Whole blood collected 4 h after the meal was subjected to an ex vivo lipopolysaccharide (LPS) challenge to evaluate cytokine secretion from blood cells. Calves were euthanized on d 25 for collection of intestinal tissue samples. Tissue samples were processed to assess FA composition by gas chromatography, histomorphology by bright-field microscopy, and gene expression of tight junction proteins, lipid metabolism enzymes, and immune molecules by real-time quantitative PCR. Data were analyzed using PROC GLIMMIX in SAS (version 9.4, SAS Institute Inc.). Growth performance and fecal consistency were unaffected. Calves fed MR with a lower ratio of n-6 to n-3 FA had 2-fold higher n-3 FA contents and 2-fold lower ratios of n-6 to n-3 FA in proximal jejunum and ileum tissues. Total urinary recovery (0-24 h relative to marker administration) and plasma concentrations of the markers were unaffected. Expression of TJP1 tended to be higher in proximal jejunum tissue and lower in ileum tissue of n-3 calves. The expression of TLR4 and TNFA tended to be higher and CD14 was higher in ileum tissue of n-3 calves. Plasma concentrations of interleukin-4 were decreased in response to the ex vivo LPS challenge in n-3 calves. Histomorphology and GIT permeability were largely unaffected by treatment. Furthermore, the inclusion of linseed and algae oil may promote inflammation, as suggested by greater concentrations of the acute-phase proteins haptoglobin and serum amyloid A postprandially, demonstrating that fat sources should be evaluated for their suitability for MR formulations. Understanding how MR composition affects dairy calf health may improve nutritional strategies on farm.
Collapse
Affiliation(s)
- A C Welboren
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - J Van Baal
- Trouw Nutrition Research and Development, PO Box 299, 3800 AG, Amersfoort, the Netherlands
| | - M H Ghaffari
- Institute of Animal Science, University of Bonn, 53111 Bonn, Germany
| | - H Sauerwein
- Institute of Animal Science, University of Bonn, 53111 Bonn, Germany
| | - J B Renaud
- London Research and Development Centre, Agriculture and Agri-Food Canada, London, ON, Canada, N5V 4T3
| | - J Martín-Tereso
- Trouw Nutrition Research and Development, PO Box 299, 3800 AG, Amersfoort, the Netherlands
| | - M A Steele
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - L N Leal
- Trouw Nutrition Research and Development, PO Box 299, 3800 AG, Amersfoort, the Netherlands.
| |
Collapse
|
10
|
Álvarez P, Ramiro-Cortijo D, Montes MT, Moreno B, Calvo MV, Liu G, Esteban Romero A, Ybarra M, Cordeiro M, Clambor Murube M, Valverde E, Sánchez-Pacheco A, Fontecha J, Gibson R, Saenz de Pipaon M. Randomized controlled trial of early arachidonic acid and docosahexaenoic acid enteral supplementation in very preterm infants. Front Pediatr 2022; 10:947221. [PMID: 36090567 PMCID: PMC9452757 DOI: 10.3389/fped.2022.947221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To evaluate changes in blood long-chain polyunsaturated fatty acid (LCPUFA) and oxylipin concentrations in very preterm infants from birth to 36 weeks' postmenstrual age (WPA) after providing an emulsified arachidonic acid (ARA):docosahexaenoic acid (DHA) supplement at two different concentrations. STUDY DESIGN This prospective, randomized trial assigned infants to receive a supplement (1) 80:40 group (80 mg/kg/day ARA and 40 mg/kg/day DHA, n = 9) or (2) 120:60 group (120 mg/kg/day ARA and 60 mg/kg/day DHA, n = 9). Infants received supplement daily from birth until 36 WPA. At baseline, 21 days of life and 36 WPA, the LCPUFAs were measured in plasma by gas chromatography/mass spectrophotometry. Additionally, LCPUFAs and oxylipins were analyzed in whole blood by ultra-high-performance liquid chromatography-tandem mass spectrometry. Furthermore, a sample of oral mucosa was obtained to analyze single-nucleotide polymorphism located in the FADS1 gene by PCR. RESULTS Gestational age was similar between groups (80:40 = 28+6 [27+3; 30+3] completed weeks+days ; 120:60 = 29+6 [27+3; 30+5] completed weeks+days , p = 0.83). At 36 WPA, the change in plasma ARA was significantly different between groups (80:40 group = 0.15 [-0.67; 0.69] %nmol, 120:60 = 1.68 [1.38; 3.16] %nmol, p = 0.031). In whole blood, the levels of ARA-derived oxylipins (5-, 8-, 9-, 11-, 15-HETE and 8,9-EET) and EPA-derived oxylipins (18-HEPE) significantly increase from baseline to 36 WPA in the 120:60 group than the 80:40 group. CONCLUSION Supplementation at high doses (120:60 mg/kg/day) increased levels of ARA, and EPA- and ARA-derived oxylipins compared to low doses (80:40 mg/kg/day). Differences were detected in EPA metabolites without a significant increase in plasma DHA.
Collapse
Affiliation(s)
- Patricia Álvarez
- Department of Neonatology, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| | - David Ramiro-Cortijo
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - María Teresa Montes
- Department of Neonatology, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| | - Bárbara Moreno
- Department of Neonatology, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| | - María V Calvo
- Food Lipid Biomarkers and Health Group, Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM), Madrid, Spain
| | - Ge Liu
- South Australian Health and Medical Research Institute, SAHMRI Women and Kids, Adelaide, SA, Australia
| | - Ana Esteban Romero
- Department of Neonatology, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta Ybarra
- Department of Neonatology, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| | - Malaika Cordeiro
- Department of Neonatology, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| | - Marina Clambor Murube
- Department of Biochemistry, Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
| | - Eva Valverde
- Department of Neonatology, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| | - Aurora Sánchez-Pacheco
- Department of Biochemistry, Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
| | - Javier Fontecha
- Food Lipid Biomarkers and Health Group, Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM), Madrid, Spain
| | - Robert Gibson
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,School of Agriculture, Food and Wine, The University of Adelaide, Adelaide, SA, Australia
| | - Miguel Saenz de Pipaon
- Department of Neonatology, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
11
|
Alshaikh BN, Reyes Loredo A, Knauff M, Momin S, Moossavi S. The Role of Dietary Fats in the Development and Prevention of Necrotizing Enterocolitis. Nutrients 2021; 14:145. [PMID: 35011027 PMCID: PMC8746672 DOI: 10.3390/nu14010145] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/21/2021] [Accepted: 12/26/2021] [Indexed: 11/18/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a significant cause of mortality and morbidity in preterm infants. The pathogenesis of NEC is not completely understood; however, intestinal immaturity and excessive immunoreactivity of intestinal mucosa to intraluminal microbes and nutrients appear to have critical roles. Dietary fats are not only the main source of energy for preterm infants, but also exert potent effects on intestinal development, intestinal microbial colonization, immune function, and inflammatory response. Preterm infants have a relatively low capacity to digest and absorb triglyceride fat. Fat may thereby accumulate in the ileum and contribute to the development of NEC by inducing oxidative stress and inflammation. Some fat components, such as long-chain polyunsaturated fatty acids (LC-PUFAs), also exert immunomodulatory roles during the early postnatal period when the immune system is rapidly developing. LC-PUFAs may have the ability to modulate the inflammatory process of NEC, particularly when the balance between n3 and n6 LC-PUFAs derivatives is maintained. Supplementation with n3 LC-PUFAs alone may have limited effect on NEC prevention. In this review, we describe how various fatty acids play different roles in the pathogenesis of NEC in preterm infants.
Collapse
Affiliation(s)
- Belal N Alshaikh
- Neonatal Nutrition and Gastroenterology Program, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2T9, Canada
| | - Adriana Reyes Loredo
- Neonatal Nutrition and Gastroenterology Program, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2T9, Canada
| | - Megan Knauff
- Nutrition Services, Alberta Health Services, Calgary, AB T2N 2T9, Canada
| | - Sarfaraz Momin
- Neonatal Nutrition and Gastroenterology Program, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2T9, Canada
| | - Shirin Moossavi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2T9, Canada
- International Microbiome Centre, Cumming School of Medicine, Health Sciences Centre, University of Calgary, Calgary, AB T2N 2T9, Canada
| |
Collapse
|
12
|
Untargeted Metabolomic Analysis of Human Milk from Mothers of Preterm Infants. Nutrients 2021; 13:nu13103604. [PMID: 34684605 PMCID: PMC8540315 DOI: 10.3390/nu13103604] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/05/2021] [Accepted: 10/09/2021] [Indexed: 12/11/2022] Open
Abstract
The application of metabolomics in neonatology offers an approach to investigate the complex relationship between nutrition and infant health. Characterization of the metabolome of human milk enables an investigation into nutrients that affect the neonatal metabolism and identification of dietary interventions for infants at risk of diseases such as necrotizing enterocolitis (NEC). In this study, we aimed to identify differences in the metabolome of breast milk of 48 mothers with preterm infants with NEC and non-NEC healthy controls. A minimum significant difference was observed in the human milk metabolome between the mothers of infants with NEC and mothers of healthy control infants. However, significant differences in the metabolome related to fatty acid metabolism, oligosaccharides, amino sugars, amino acids, vitamins and oxidative stress-related metabolites were observed when comparing milk from mothers with control infants of ≤1.0 kg birth weight and >1.5 kg birth weight. Understanding the functional biological features of mothers’ milk that may modulate infant health is important in the future of tailored nutrition and care of the preterm newborn.
Collapse
|
13
|
Lau E, Lee C, Li B, Pierro A. Endoplasmic reticulum stress in the acute intestinal epithelial injury of necrotizing enterocolitis. Pediatr Surg Int 2021; 37:1151-1160. [PMID: 34117913 DOI: 10.1007/s00383-021-04929-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/10/2021] [Indexed: 03/07/2023]
Abstract
Endoplasmic reticulum (ER) is a dynamic organelle that has many functions including protein synthesis, lipid synthesis, and calcium metabolism. Any perturbation in the ER such as accumulation of unfolded or misfolded proteins in the ER lumen causes ER stress. ER stress has been implicated in many intestinal inflammatory diseases. However, the role of ER stress in acute intestinal epithelial injuries such as necrotizing enterocolitis in preterm neonates, remains incompletely understood. In this review, we introduce ER structure, functions and summarize the intracellular signaling pathways involved in unfolded protein response (UPR), a survival mechanism in which cells exert an adaptive function to restore homeostasis in the ER. However, intense and prolonged ER stress induces apoptotic response which results in apoptotic cell death. We also discuss and highlight recent advances that have improved our understanding of the molecular mechanisms that regulate the ER stress in acute intestinal epithelial injuries such as necrotizing enterocolitis (NEC). We focus on the role of ER stress in influencing gut homeostasis in the neonatal period and on the potential therapeutic interventions to alleviate ER stress-induced cell death in NEC.
Collapse
Affiliation(s)
- Ethan Lau
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, 1526-555 University Ave, Toronto, ON, M5G 1X8, Canada
| | - Carol Lee
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, 1526-555 University Ave, Toronto, ON, M5G 1X8, Canada
| | - Bo Li
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, 1526-555 University Ave, Toronto, ON, M5G 1X8, Canada
| | - Agostino Pierro
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, 1526-555 University Ave, Toronto, ON, M5G 1X8, Canada.
| |
Collapse
|
14
|
de Lange IH, van Gorp C, Eeftinck Schattenkerk LD, van Gemert WG, Derikx JPM, Wolfs TGAM. Enteral Feeding Interventions in the Prevention of Necrotizing Enterocolitis: A Systematic Review of Experimental and Clinical Studies. Nutrients 2021; 13:1726. [PMID: 34069699 PMCID: PMC8161173 DOI: 10.3390/nu13051726] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 12/11/2022] Open
Abstract
Necrotizing enterocolitis (NEC), which is characterized by severe intestinal inflammation and in advanced stages necrosis, is a gastrointestinal emergency in the neonate with high mortality and morbidity. Despite advancing medical care, effective prevention strategies remain sparse. Factors contributing to the complex pathogenesis of NEC include immaturity of the intestinal immune defense, barrier function, motility and local circulatory regulation and abnormal microbial colonization. Interestingly, enteral feeding is regarded as an important modifiable factor influencing NEC pathogenesis. Moreover, breast milk, which forms the currently most effective prevention strategy, contains many bioactive components that are known to support neonatal immune development and promote healthy gut colonization. This systematic review describes the effect of different enteral feeding interventions on the prevention of NEC incidence and severity and the effect on pathophysiological mechanisms of NEC, in both experimental NEC models and clinical NEC. Besides, pathophysiological mechanisms involved in human NEC development are briefly described to give context for the findings of altered pathophysiological mechanisms of NEC by enteral feeding interventions.
Collapse
Affiliation(s)
- Ilse H. de Lange
- European Surgical Center Aachen/Maastricht, Department of Pediatric Surgery, School for Nutrition, Toxicology and Metabolism (NUTRIM), 6202 AZ Maastricht, The Netherlands; (I.H.d.L.); (W.G.v.G.)
- Department of Surgery, School for Nutrition, Toxicology and Metabolism (NUTRIM), Maastricht University, 6202 AZ Maastricht, The Netherlands
- Department of Pediatrics, School of Oncology and Developmental Biology (GROW), Maastricht University, 6202 AZ Maastricht, The Netherlands;
| | - Charlotte van Gorp
- Department of Pediatrics, School of Oncology and Developmental Biology (GROW), Maastricht University, 6202 AZ Maastricht, The Netherlands;
| | - Laurens D. Eeftinck Schattenkerk
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, 1105 AZ Amsterdam, The Netherlands; (L.D.E.S.); (J.P.M.D.)
| | - Wim G. van Gemert
- European Surgical Center Aachen/Maastricht, Department of Pediatric Surgery, School for Nutrition, Toxicology and Metabolism (NUTRIM), 6202 AZ Maastricht, The Netherlands; (I.H.d.L.); (W.G.v.G.)
- Department of Surgery, School for Nutrition, Toxicology and Metabolism (NUTRIM), Maastricht University, 6202 AZ Maastricht, The Netherlands
| | - Joep P. M. Derikx
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, 1105 AZ Amsterdam, The Netherlands; (L.D.E.S.); (J.P.M.D.)
| | - Tim G. A. M. Wolfs
- Department of Pediatrics, School of Oncology and Developmental Biology (GROW), Maastricht University, 6202 AZ Maastricht, The Netherlands;
- Department of Biomedical Engineering (BMT), School for Cardiovascular Diseases (CARIM), Maastricht University, 6202 AZ Maastricht, The Netherlands
| |
Collapse
|
15
|
Quitadamo PA, Comegna L, Cristalli P. Anti-Infective, Anti-Inflammatory, and Immunomodulatory Properties of Breast Milk Factors for the Protection of Infants in the Pandemic From COVID-19. Front Public Health 2021; 8:589736. [PMID: 33738273 PMCID: PMC7960784 DOI: 10.3389/fpubh.2020.589736] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
COVID-19 pandemic since the end of 2019 spreads worldwide, counting millions of victims. The viral invasion, systemic inflammation, and consequent organ failure are the gravest features of coronavirus disease 2019 (COVID-19), and they are associated with a high mortality rate. The aim of this study is to evaluate the role of breast milk in the COVID-19 pandemic, analyzing its antiviral, anti-inflammatory, and immunoregulatory effects due to its bioactive components, so numerous and important for the protection of infants. The study tried to demonstrate that all the components of human milk are capable of performing functions on all the pathogenic events recognized and described in COVID-19 disease. Those human milk factors are well-tolerated and practically free of side effects, so breast milk should become a research topic to discover therapies even in this epidemic. In the first part, the mechanisms of protection and defense of the breast milk elements will be delineated; in the second section, it will describe the human milk effects in viral infections and it will be hypothesized how the known mechanisms could act in COVID infection.
Collapse
Affiliation(s)
- Pasqua Anna Quitadamo
- NICU “Casa Sollievo della Sofferenza” Foundation, Scientific Research and Care Institute, San Giovanni Rotondo, Italy
| | | | | |
Collapse
|
16
|
Lipid Composition, Digestion, and Absorption Differences among Neonatal Feeding Strategies: Potential Implications for Intestinal Inflammation in Preterm Infants. Nutrients 2021; 13:nu13020550. [PMID: 33567518 PMCID: PMC7914900 DOI: 10.3390/nu13020550] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/18/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a significant cause of morbidity and mortality in the neonatal population. Formula feeding is among the many risk factors for developing the condition, a practice often required in the cohort most often afflicted with NEC, preterm infants. While the virtues of many bioactive components of breast milk have been extolled, the ability to digest and assimilate the nutritional components of breast milk is often overlooked. The structure of formula differs from that of breast milk, both in lipid composition and chemical configuration. In addition, formula lacks a critical digestive enzyme produced by the mammary gland, bile salt-stimulated lipase (BSSL). The gastrointestinal system of premature infants is often incapable of secreting sufficient pancreatic enzymes for fat digestion, and pasteurization of donor milk (DM) has been shown to inactivate BSSL, among other important compounds. Incompletely digested lipids may oxidize and accumulate in the distal gut. These lipid fragments are thought to induce intestinal inflammation in the neonate, potentially hastening the development of diseases such as NEC. In this review, differences in breast milk, pasteurized DM, and formula lipids are highlighted, with a focus on the ability of those lipids to be digested and subsequently absorbed by neonates, especially those born prematurely and at risk for NEC.
Collapse
|
17
|
Durkin LA, Childs CE, Calder PC. Omega-3 Polyunsaturated Fatty Acids and the Intestinal Epithelium-A Review. Foods 2021; 10:foods10010199. [PMID: 33478161 PMCID: PMC7835870 DOI: 10.3390/foods10010199] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Epithelial cells (enterocytes) form part of the intestinal barrier, the largest human interface between the internal and external environments, and responsible for maintaining regulated intestinal absorption and immunological control. Under inflammatory conditions, the intestinal barrier and its component enterocytes become inflamed, leading to changes in barrier histology, permeability, and chemical mediator production. Omega-3 (ω-3) polyunsaturated fatty acids (PUFAs) can influence the inflammatory state of a range of cell types, including endothelial cells, monocytes, and macrophages. This review aims to assess the current literature detailing the effects of ω-3 PUFAs on epithelial cells. Marine-derived ω-3 PUFAs, eicosapentaenoic acid and docosahexaenoic acid, as well as plant-derived alpha-linolenic acid, are incorporated into intestinal epithelial cell membranes, prevent changes to epithelial permeability, inhibit the production of pro-inflammatory cytokines and eicosanoids and induce the production of anti-inflammatory eicosanoids and docosanoids. Altered inflammatory markers have been attributed to changes in activity and/or expression of proteins involved in inflammatory signalling including nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), peroxisome proliferator activated receptor (PPAR) α and γ, G-protein coupled receptor (GPR) 120 and cyclooxygenase (COX)-2. Effective doses for each ω-3 PUFA are difficult to determine due to inconsistencies in dose and time of exposure between different in vitro models and between in vivo and in vitro models. Further research is needed to determine the anti-inflammatory potential of less-studied ω-3 PUFAs, including docosapentaenoic acid and stearidonic acid.
Collapse
Affiliation(s)
- Luke A. Durkin
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (C.E.C.); (P.C.C.)
- Correspondence:
| | - Caroline E. Childs
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (C.E.C.); (P.C.C.)
- Institute of Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Philip C. Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (C.E.C.); (P.C.C.)
- Institute of Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
18
|
Kerry RG, Das G, Golla U, Del Pilar Rodriguez-Torres M, Shin H, Patra JK. Engineered probiotic and prebiotic nutraceutical supplementations in combating non-communicable disorders: A review. Curr Pharm Biotechnol 2020; 23:72-97. [PMID: 33050862 DOI: 10.2174/1389201021666201013153142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 11/22/2022]
Abstract
Nutritional supplementations are a form of nutrition sources that may help in improving health complexities throughout the life span of a person. Under the umbrella of food supplementations, nutraceuticals are products extracted from edible sources that provide medical benefits along with primary nutritional value, these can be considered as functional foods. These nutraceutical supplementations are also evidenced in altering the commensal gut microbiota and help to prevent or fight against chronic non-communicable degenerative diseases in adults including neurological disorders (Autism Spectrum Disorder [ASD], Parkinson's disease [PD] and Multiple sclerosis [MS]) and metabolic disorder (Type-II Diabetes, Obesity and non-alcoholic fatty liver disease). Even the complexities of preterm babies like extra-uterine growth restriction, necrotizing enterocolitis, infant eczema and allergy (during pregnancy) and bronchopulmonary dysplasia, etc. could also be lessened up by providing proper nutrition. Molecular perceptive of inflammatory and apoptotic modulators regulating the pathogenesis of these health risks, their control and management by probiotics and prebiotics could further emphasize the scientific overview of their utility. The pivotal role of nutraceutical supplementations in regulating or modulating molecular pathways coupled with the above mentioned non-communicable diseases are briefly described. Lastly, an overall introduction to the sophisticated genome-editing techniques and advanced delivery systems in therapeutic activities applicable under these health risks are also emphasized in this paper.
Collapse
Affiliation(s)
| | - Gitishree Das
- Research Institute of Biotechnology & Medical Converged Science, Dongguk University-Seoul, Goyangsi 10326. Korea
| | - Upendarrao Golla
- Division of Hematology and Oncology, Penn State College of Medicine, Hershey, PA 17033. United States
| | - Maria Del Pilar Rodriguez-Torres
- Laboratorio de Ondas de Choque (LOCH), Centro de Física Aplicada y Tecnología Avanzada (CFATA), Universidad Nacional Autónoma de México, Campus UNAM Juriquilla Boulevard Juriquilla no. 3001, Santiago de Querétaro, Qro., C.P. 76230. Mexico
| | - HanSeung Shin
- Department of Food Science and Biotechnology, Dongguk University-Seoul, Goyangsi 10326. Korea
| | - Jayanta Kumar Patra
- Research Institute of Biotechnology & Medical Converged Science, Dongguk University-Seoul, Goyangsi 10326. Korea
| |
Collapse
|
19
|
Abstract
Mother's own human milk is the best nutrition for infants, especially preterm very-low-birth-weight (VLBW) (≤1,500 g) infants, because of its immune-modulatory constituents that strengthen the infant's host defense, provide protection against infections, and decrease the risk of necrotizing enterocolitis (NEC). When mother's own milk is unavailable or insufficient, donor human milk is considered the best alternative, especially for preterm VLBW infants. However, to assure biological safety, donor milk must be pasteurized. This results in partial or complete inactivation of some of the immunomodulatory constituents of human milk, which confer host defense. This review summarizes the current evidence regarding the effects of pasteurization on the different immunological constituents of donor milk, and their clinical significance, especially in relation to prevention of NEC.
Collapse
Affiliation(s)
- Arieh Riskin
- Department of Neonatology, Bnai Zion Medical Center, Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
20
|
Zhu X, Cui N, Yu L, Cheng P, Cui M, Zhu X, Wang J. Potential role of endoplasmic reticulum stress is involved in the protection of fish oil on neonatal rats with necrotizing enterocolitis. Sci Rep 2020; 10:6448. [PMID: 32296092 PMCID: PMC7160196 DOI: 10.1038/s41598-020-63309-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 03/09/2020] [Indexed: 11/09/2022] Open
Abstract
Neonatal necrotizing enterocolitis (NEC) is a serious gastrointestinal disease with high death rate in premature infants. Fish oil (FO) and its constituents have been shown to ameliorate intestinal inflammation and mucosal damage. However, the underlying mechanism of action is not known. In the present study, we divided Sprague-Dawley rats into three groups: control group, NEC model group, and FO pre-feeding+NEC model group. Briefly, one week before NEC modeling, in addition to being fed with milk, the FO pre-feeding+NEC modeling group was fed with FO, the NEC group was fed with saline, and the control group was only inserted a gastric-tube for 7 days. Subsequently, histological assay, Western blot, and ELISA were performed. Pretreatment with FO attenuated the NEC symptoms, alleviated intestinal pathological injury, and decreased the expressions of pro-inflammatory cytokines such as interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). Furthermore, pretreatment with FO reduced the expressions of endoplasmic reticulum stress (ERS) related proteins, caspase-12, and glucose-regulated protein 78 (GRP78). In addition, intestinal histopathological scores showed a significant positive correlation with intestinal expressions of IL-6, TNF-α, and caspase-12. Collectively, these results indicate that ERS pathway might be involved in the effect of FO in alleviating intestinal mucosal inflammation and injury in rats with NEC.
Collapse
Affiliation(s)
- Xiaoli Zhu
- Department of Intervention, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Ningxun Cui
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Lingling Yu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Ping Cheng
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Mingling Cui
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Xueping Zhu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China.
| | - Jian Wang
- Department of Neonatology Surgery, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu, China.
| |
Collapse
|
21
|
Prolyl hydroxylase 2 silencing enhances the paracrine effects of mesenchymal stem cells on necrotizing enterocolitis in an NF-κB-dependent mechanism. Cell Death Dis 2020; 11:188. [PMID: 32179740 PMCID: PMC7075868 DOI: 10.1038/s41419-020-2378-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
Abstract
Treatment options for necrotizing enterocolitis (NEC) remain inadequate. Here we examined if and how prolyl hydroxylase 2 (PHD2) silencing enhances the paracrine effects of bone-marrow-derived mesenchymal stem cells (BM-MSCs) on NEC. In this study, BM-MSCs were transduced with lentiviruses containing GFP (GFP-MSC) or shPHD2-GFP constructs (PHDMSC), followed by intraperitoneal injection of the PHDMSC-conditioned medium (PHDMSC-CM) or the GFP-MSC-conditioned medium (MSC-CM) into a rat pup model of NEC. Our results showed that systemic infusion of PHDMSC-CM, but not MSC-CM, significantly improved intestinal damage and survival of NEC rats. Such benefits may involve the modulation of epithelial regeneration and inflammation, as indicated by the regeneration of intestinal epithelial/stem cells, the regulation of Treg cells function and pro-/anti-inflammatory cytokine balance. The mechanism for the superior paracrine efficacy of PHDMSC is related to a higher release of pivotal factor IGF-1 and TGF-β2. NF-κB activation was induced by PHD2 silencing to induce IGF-1 and TGF-β2 secretion via binding to IGF-1 and TGF-β2 gene promoter. Our work indicated that PHD2 silencing enhanced the paracrine effect of BM-MSCs on NEC via the NF-κB-dependent mechanism which may be a novel strategy for stem cell therapy on NEC.
Collapse
|
22
|
Thai JD, Gregory KE. Bioactive Factors in Human Breast Milk Attenuate Intestinal Inflammation during Early Life. Nutrients 2020; 12:E581. [PMID: 32102231 PMCID: PMC7071406 DOI: 10.3390/nu12020581] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/15/2020] [Accepted: 02/16/2020] [Indexed: 12/14/2022] Open
Abstract
Human breast milk is well known as the ideal source of nutrition during early life, ensuring optimal growth during infancy and early childhood. Breast milk is also the source of many unique and dynamic bioactive components that play a key role in the development of the immune system. These bioactive components include essential microbes, human milk oligosaccharides (HMOs), immunoglobulins, lactoferrin and dietary polyunsaturated fatty acids. These factors all interact with intestinal commensal bacteria and/or immune cells, playing a critical role in establishment of the intestinal microbiome and ultimately influencing intestinal inflammation and gut health during early life. Exposure to breast milk has been associated with a decreased incidence and severity of necrotizing enterocolitis (NEC), a devastating disease characterized by overwhelming intestinal inflammation and high morbidity among preterm infants. For this reason, breast milk is considered a protective factor against NEC and aberrant intestinal inflammation common in preterm infants. In this review, we will describe the key microbial, immunological, and metabolic components of breast milk that have been shown to play a role in the mechanisms of intestinal inflammation and/or NEC prevention.
Collapse
Affiliation(s)
- Julie D. Thai
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Katherine E. Gregory
- Department of Pediatric Newborn Medicine, Department of Nursing, Brigham and Women’s Hospital, Boston, MA 02115, USA;
| |
Collapse
|
23
|
Ramiro-Cortijo D, Singh P, Liu Y, Medina-Morales E, Yakah W, Freedman SD, Martin CR. Breast Milk Lipids and Fatty Acids in Regulating Neonatal Intestinal Development and Protecting against Intestinal Injury. Nutrients 2020; 12:E534. [PMID: 32092925 PMCID: PMC7071444 DOI: 10.3390/nu12020534] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/14/2020] [Accepted: 02/16/2020] [Indexed: 12/13/2022] Open
Abstract
Human breast milk is the optimal source of nutrition for infant growth and development. Breast milk fats and their downstream derivatives of fatty acids and fatty acid-derived terminal mediators not only provide an energy source but also are important regulators of development, immune function, and metabolism. The composition of the lipids and fatty acids determines the nutritional and physicochemical properties of human milk fat. Essential fatty acids, including long-chain polyunsaturated fatty acids (LCPUFAs) and specialized pro-resolving mediators, are critical for growth, organogenesis, and regulation of inflammation. Combined data including in vitro, in vivo, and human cohort studies support the beneficial effects of human breast milk in intestinal development and in reducing the risk of intestinal injury. Human milk has been shown to reduce the occurrence of necrotizing enterocolitis (NEC), a common gastrointestinal disease in preterm infants. Preterm infants fed human breast milk are less likely to develop NEC compared to preterm infants receiving infant formula. Intestinal development and its physiological functions are highly adaptive to changes in nutritional status influencing the susceptibility towards intestinal injury in response to pathological challenges. In this review, we focus on lipids and fatty acids present in breast milk and their impact on neonatal gut development and the risk of disease.
Collapse
Affiliation(s)
- David Ramiro-Cortijo
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; (D.R.-C.); (P.S.); (Y.L.); (E.M.-M.); (S.D.F.)
| | - Pratibha Singh
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; (D.R.-C.); (P.S.); (Y.L.); (E.M.-M.); (S.D.F.)
| | - Yan Liu
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; (D.R.-C.); (P.S.); (Y.L.); (E.M.-M.); (S.D.F.)
| | - Esli Medina-Morales
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; (D.R.-C.); (P.S.); (Y.L.); (E.M.-M.); (S.D.F.)
| | - William Yakah
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA;
| | - Steven D. Freedman
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; (D.R.-C.); (P.S.); (Y.L.); (E.M.-M.); (S.D.F.)
- Division of Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Camilia R. Martin
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA;
- Division of Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| |
Collapse
|
24
|
Abstract
Necrotising enterocolitis (NEC) is a devastating disease that typically affects formula-fed premature infants, suggesting that dietary components may influence disease pathogenesis. TAG are the major fat components of infant formula, and their digestion requires pancreatic lipases, which may be naturally deficient in premature neonates. We hypothesise that NEC develops partly from the accumulation of incompletely digested long-chain TAG-containing unsaturated fatty acids within the intestinal epithelial cells, leading to oxidative stress and enterocyte damage. We further hypothesise that the administration of a formula that contains reduced TAG ('pre-digested fat') that do not require lipase action may reduce NEC severity. To test these hypotheses, we induced NEC in neonatal mice using three different fat formulations, namely 'standard fat', 'pre-digested fat' or 'very low fat', and determined that mice fed 'standard fat' developed severe NEC, which was significantly reduced in mice fed 'pre-digested fat' or 'very low fat'. The expression level of the critical fat-digesting enzyme carboxyl ester lipase was significantly lower in the newborn compared with older pups, leading to impaired fat digestion. The accumulation of mal-digested fat resulted in the significant accumulation of fat droplets within the intestinal epithelium of the distal ileum, resulting in the generation of reactive oxygen species and intestinal inflammation. Strikingly, these changes were prevented in pups fed 'pre-digested fat' or 'very low fat' formulas. These findings suggest that nutritional formula containing a pre-digested fat system may overcome the natural lipase deficiency of the premature gut, and serve as a novel approach to prevent NEC.
Collapse
|
25
|
Frost BL, Caplan MS. Can Fish Oil Reduce the Incidence of Necrotizing Enterocolitis by Altering the Inflammatory Response? Clin Perinatol 2019; 46:65-75. [PMID: 30771820 DOI: 10.1016/j.clp.2018.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Necrotizing enterocolitis (NEC) is a devastating bowel necrosis that predominantly affects preterm infants and is characterized by an imbalance toward a proinflammatory state. Fish oil or omega-3 long-chain polyunsaturated fatty acids have the potential to modulate inflammation. In this article, the authors examine the evidence in support of fish oil supplementation to alter the inflammatory response and potentially reduce the risk of NEC.
Collapse
Affiliation(s)
- Brandy L Frost
- Department of Pediatrics, NorthShore University HealthSystem, University of Chicago Pritzker School of Medicine, 2650 Ridge Avenue, Walgreen Building Suite 1505, Evanston, IL 60201, USA.
| | - Michael S Caplan
- Department of Pediatrics, NorthShore University HealthSystem, University of Chicago Pritzker School of Medicine, 2650 Ridge Avenue, Walgreen Building Suite 1505, Evanston, IL 60201, USA
| |
Collapse
|
26
|
Teresa C, Antonella D, de Ville de Goyet Jean. New Nutritional and Therapeutical Strategies of NEC. Curr Pediatr Rev 2019; 15:92-105. [PMID: 30868956 DOI: 10.2174/1573396315666190313164753] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/09/2018] [Accepted: 03/06/2019] [Indexed: 11/22/2022]
Abstract
Necrotizing enterocolitis (NEC) is an acquired severe disease of the digestive system affecting mostly premature babies, possibly fatal and frequently associated to systemic complications. Because of the severity of this condition and the possible long-term consequences on the child's development, many studies have aimed at preventing the occurrence of the primary events at the level of the bowel wall (ischemia and necrosis followed by sepsis) by modifying or manipulating the diet (breast milk versus formula) and/or the feeding pattern (time for initiation after birth, continuous versus bolus feeding, modulation of intake according clinical events). Feeding have been investigated so far in order to prevent NEC. However, currently well-established and shared clinical nutritional practices are not available in preventing NEC. Nutritional and surgical treatments of NEC are instead well defined. In selected cases surgery is a therapeutic option of NEC, requiring sometimes partial intestinal resection responsible for short bowel syndrome. In this paper we will investigate the available options for treating NEC according to the Walsh and Kliegman classification, focusing on feeding practices in managing short bowel syndrome that can complicate NEC. We will also analyze the proposed ways of preventing NEC.
Collapse
Affiliation(s)
- Capriati Teresa
- Artificial Nutrition in Pediatric Children's Hospital, Bambino Gesu, Rome, Italy
| | - Diamanti Antonella
- Artificial Nutrition in Pediatric Children's Hospital, Bambino Gesu, Rome, Italy
| | - de Ville de Goyet Jean
- Pediatric Department for the Treatment and Study of abdominal Disease and Abdominal Transplants, ISMETT-UPMC, Palermo, Italy
| |
Collapse
|
27
|
Lenfestey MW, Neu J. Gastrointestinal Development: Implications for Management of Preterm and Term Infants. Gastroenterol Clin North Am 2018; 47:773-791. [PMID: 30337032 DOI: 10.1016/j.gtc.2018.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The gastrointestinal (GI) system provides digestive, absorptive, neuroendocrine, and immunologic functions to support overall health. If normal development is interrupted, a variety of complications and disease can arise. This article explores normal development of the GI tract and specific clinical challenges pertinent to preterm and term infants. Specific topics include abnormal motility, gastroesophageal reflux, current feeding recommendations for preterm infants, effects of parenteral nutrition, and the relationship between the GI tract and the immune system.
Collapse
Affiliation(s)
- Mary W Lenfestey
- Department of Pediatrics, University of Florida, PO Box 100296, Gainesville, FL 32610, USA
| | - Josef Neu
- Department of Pediatrics, University of Florida, 6516 Southwest 93rd Avenue, Gainesville, FL 32610, USA.
| |
Collapse
|
28
|
Knoop KA, Holtz LR, Newberry RD. Inherited nongenetic influences on the gut microbiome and immune system. Birth Defects Res 2018; 110:1494-1503. [PMID: 30576093 PMCID: PMC8759455 DOI: 10.1002/bdr2.1436] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/16/2018] [Indexed: 12/18/2022]
Abstract
The gut microbiome and the immune system codevelop around the time of birth, well after genetic information has been passed from the parents to the offspring. Each of these "organ systems" displays plasticity. The immune system can mount highly specific adaptive responses to newly encountered antigens, and the gut microbiota is affected by changes in the environment. Despite this plasticity, there is a growing appreciation that these organ systems, once established, are remarkably stable. In health, the immune system rapidly mounts responses to infections, and once cleared, resolves inflammatory responses to return to homeostasis. However, a skewed immune system, such as seen in allergy, does not easily return to homeostasis. Allergic responses are often seen to multiple antigens. Likewise, a dysbiotic gut microbiota is seen in multiple diseases. Attempts to reset the gut microbiota as a therapy for disease have met with varied success. Therefore, how these codeveloping "organ systems" become established is a central question relevant to our overall health. Recent observations suggest that maternal factors encountered both in utero and after birth can directly or indirectly impact the development of the offspring's gut microbiome and immune system. Here, we discuss how these nongenetic maternal influences can have long-term effects on the progeny's health.
Collapse
Affiliation(s)
- Kathryn A. Knoop
- Department of Medicine, Washington University School of Medicine in Saint Louis, MO 63110
| | - Lori R. Holtz
- Department of Pediatrics, Washington University School of Medicine in Saint Louis, MO 63110
| | - Rodney D. Newberry
- Department of Medicine, Washington University School of Medicine in Saint Louis, MO 63110
| |
Collapse
|
29
|
Autran CA, Kellman BP, Kim JH, Asztalos E, Blood AB, Spence ECH, Patel AL, Hou J, Lewis NE, Bode L. Human milk oligosaccharide composition predicts risk of necrotising enterocolitis in preterm infants. Gut 2018; 67:1064-1070. [PMID: 28381523 DOI: 10.1136/gutjnl-2016-312819] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 03/07/2017] [Accepted: 03/08/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Necrotising enterocolitis (NEC) is one of the most common and often fatal intestinal disorders in preterm infants. Markers to identify at-risk infants as well as therapies to prevent and treat NEC are limited and urgently needed. NEC incidence is significantly lower in breast-fed compared with formula-fed infants. Infant formula lacks human milk oligosaccharides (HMO), such as disialyllacto-N-tetraose (DSLNT), which prevents NEC in neonatal rats. However, it is unknown if DSLNT also protects human preterm infants. DESIGN We conducted a multicentre clinical cohort study and recruited 200 mothers and their very low birthweight infants that were predominantly human milk-fed. We analysed HMO composition in breast milk fed to infants over the first 28 days post partum, matched each NEC case with five controls and used logistic regression and generalised estimating equation to test the hypothesis that infants who develop NEC receive milk with less DSLNT than infants who do not develop NEC. RESULTS Eight infants in the cohort developed NEC (Bell stage 2 or 3). DSLNT concentrations were significantly lower in almost all milk samples in NEC cases compared with controls, and its abundance could identify NEC cases prior to onset. Aggregate assessment of DSLNT over multiple days enhanced the separation of NEC cases and control subjects. CONCLUSIONS DSLNT content in breast milk is a potential non-invasive marker to identify infants at risk of developing NEC, and screen high-risk donor milk. In addition, DSLNT could serve as a natural template to develop novel therapeutics against this devastating disorder.
Collapse
Affiliation(s)
- Chloe A Autran
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
| | - Benjamin P Kellman
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA.,Bioinformatics and Systems Biology Program, University of California, La Jolla, California, USA
| | - Jae H Kim
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
| | - Elizabeth Asztalos
- Department of Newborn & Developmental Pediatrics, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Arlin B Blood
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, California, USA
| | | | - Aloka L Patel
- Rush University Medical Center, Chicago, Illinois, USA
| | - Jiayi Hou
- Clinical & Translational Research Institute, University of California San Diego, La Jolla, California, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA.,Bioinformatics and Systems Biology Program, University of California, La Jolla, California, USA.,Novo Nordisk Foundation Center for Biosustainability at the University of California San Diego School of Medicine, La Jolla, California, USA
| | - Lars Bode
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
| |
Collapse
|
30
|
Fang C, Xie L, Liu C, Fu C, Ye W, Liu H, Zhang B. Berberine ameliorates neonatal necrotizing enterocolitis by activating the phosphoinositide 3-kinase/protein kinase B signaling pathway. Exp Ther Med 2018; 15:3530-3536. [PMID: 29545879 DOI: 10.3892/etm.2018.5858] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 08/22/2017] [Indexed: 01/05/2023] Open
Abstract
Neonatal necrotizing enterocolitis (NEC) is a severe acquired disease that predominantly affects the small intestine of neonates. NEC is caused by a combination of metabolic products, dysfunctions of the blood vessels, mucus and other unknown factors. Berberine may induce beneficial effects on necrotic and cardiovascular diseases due to its anti-inflammatory and anti-apoptotic effects on epithelial cells. In the present study, the therapeutic effects of berberine were investigated and the potential mechanisms by which it functions within a neonatal NEC mouse model were analyzed. Inflammation and levels of associated factors were measured in the serum of mice with NEC prior to and following treatment with berberine. Apoptotic rates in epithelial cells were analyzed following treatment with berberine. The expression of genes associated with apoptosis and apoptosis signaling were determined in epithelial cells in the small intestines of mice with NEC following treatment with berberine. The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway was investigated in epithelial cells isolated from mice following treatment with either berberine or PBS. Histology and immunohistochemistry were used to determine the area of infarction and apoptosis. Body weight and food intake were measured to evaluate the physical effects of berberine on mice with NEC. The results indicated that berberine attenuated the inflammation caused by NEC in mice after 10 days of treatment. The apoptosis rate of epithelial cells isolated from experimental mice was decreased following berberine treatment. Western blot analysis indicated that the expression of the anti-apoptotic genes c-Myc and p53 were upregulated by berberine, whereas caspase-3 and -9 levels were downregulated in epithelial cells following treatment with berberine. In addition, the expression and phosphorylation levels of PI3K and AKT were downregulated in epithelial cells following treatment with berberine. An in vitro assay indicated that treatment with PI3K alone increased the expression of AKT and promoted the apoptosis of epithelial cells. Treatment with berberine markedly increased epidermal growth factor (EGF) and Bcl-2 expression levels, the activity of epithelial cells and decreased the infarction area of the small intestine. Accordingly, the body weight and food intake of mice with NEC were increased following berberine treatment. Therefore, the results of the present study demonstrate that berberine inhibits inflammation and apoptosis via the PI3K/AKT signaling pathway and may therefore attenuate the progression of NEC. These results suggest that berberine may be a potential therapeutic agent for the treatment of patients with NEC.
Collapse
Affiliation(s)
- Chengzhi Fang
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Lili Xie
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Chunmei Liu
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Chunhua Fu
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei Ye
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hong Liu
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Binghong Zhang
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
31
|
Robinson DT, Palac HL, Baillif V, Van Goethem E, Dubourdeau M, Van Horn L, Martin CR. Long chain fatty acids and related pro-inflammatory, specialized pro-resolving lipid mediators and their intermediates in preterm human milk during the first month of lactation. Prostaglandins Leukot Essent Fatty Acids 2017; 121:1-6. [PMID: 28651692 DOI: 10.1016/j.plefa.2017.05.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 04/10/2017] [Accepted: 05/11/2017] [Indexed: 01/08/2023]
Abstract
This study aimed to measure longitudinal quantities of the long chain fatty acids, their biologically active terminal metabolites and related intermediates (also called oxylipins) in preterm human milk expressed during the first month of lactation. In a prospective cohort, breast milk was collected throughout the first month of lactation in 30 women who delivered preterm infants. Eighteen bioactive lipids and their intermediates were quantified via solid phase extraction and LC-MS/MS. Analysis by GC-FID quantified the fatty acid precursors. Arachidonic acid (ARA) and docosahexaenoic acid (DHA) milk concentrations significantly declined throughout the first month. Oxylipin concentrations did not change during lactation. Positive associations existed between ARA and thromboxane B2, eicosapentaenoic acid and 18-hydroxyeicosapentaenoic acid, and between DHA and PDX and 14- and 17-hydroxydocosahexaenoic acids. DHA concentrations were 1.5 times higher and 14-HDHA was 1.7 times higher in milk from women taking DHA supplements. This investigation showed conditionally essential fatty acids, ARA and DHA, decreased in preterm milk, suggesting a need to supplement their intake for the breast milk-fed preterm infant. Positive associations between parent fatty acids, bioactive lipids and intermediates, as well as sensitivity of milk to maternal fatty acid intake, support consideration of a comprehensive approach to providing fatty acids for preterm infants through both maternal and infant supplementation.
Collapse
Affiliation(s)
- D T Robinson
- Department of Pediatrics, Division of Neonatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - H L Palac
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | | | | | - L Van Horn
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - C R Martin
- Department of Neonatology and Division of Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
32
|
Zhou P, Li Y, Ma LY, Lin HC. The Role of Immunonutrients in the Prevention of Necrotizing Enterocolitis in Preterm Very Low Birth Weight Infants. Nutrients 2015; 7:7256-70. [PMID: 26343718 PMCID: PMC4586529 DOI: 10.3390/nu7095334] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/19/2015] [Accepted: 08/20/2015] [Indexed: 12/13/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a critical intestinal emergency condition, which mainly occurs in preterm very low birth weight (PVLBW) infants. Despite remarkable advances in the care of PVLBW infants, with considerable improvement of the survival rate in recent decades, the incidence of NEC and NEC-related mortality have not declined accordingly. The fast progression from nonspecific signs to extensive necrosis also makes primary prevention the first priority. Recently, increasing evidence has indicated the important role of several nutrients in primary prevention of NEC. Therefore, the aim of this review is to summarize some potential immunomodulatory nutrients in the prevention of NEC, including bovine colostrum, probiotics, prebiotics (e.g., human milk oligosaccharides), long chain polyunsaturated fatty acids, and amino acids (glutamine, cysteine and N-acetylcysteine, l-arginine and l-citrulline). Based on current research evidence, probiotics are the most documented effective method to prevent NEC, while others still require further investigation in animal studies and clinical randomized controlled trials.
Collapse
Affiliation(s)
- Ping Zhou
- Department of Neonatology, Bao'an Maternal and Child Health Hospital, Shenzhen 518133, Guangdong, China.
| | - Yanqi Li
- Comparative Pediatrics and Nutrition, University of Copenhagen, Frederiksberg DK-1870, Denmark.
| | - Li-Ya Ma
- Department of Neonatology, Bao'an Maternal and Child Health Hospital, Shenzhen 518133, Guangdong, China.
| | - Hung-Chih Lin
- Children's Hospital of China Medical University, No. 2 Yuh Der Road, Taichung 404, Taiwan.
- School of Chinese Medicine, China Medical University, No. 91 Hsueh-Shih Road, Taichung 404, Taiwan.
| |
Collapse
|
33
|
Støy ACF, Østergaard MV, Sangild PT. Amniotic Fluid and Colostrum as Potential Diets in the Critical Care of Preterm Infants. DIET AND NUTRITION IN CRITICAL CARE 2015:1109-1121. [DOI: 10.1007/978-1-4614-7836-2_131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
34
|
Linking fat intake, the intestinal microbiome, and necrotizing enterocolitis in premature infants. Pediatr Res 2015; 77:121-6. [PMID: 25303279 DOI: 10.1038/pr.2014.155] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 05/04/2014] [Indexed: 12/12/2022]
Abstract
Components of diet, including the total amounts and specific types of fat, affect the composition of the intestinal microbiome in both animal models and cohort studies of humans. Amounts of total fat and specific fatty acids (FA) are some of the most variable nutritional components of breast milk. Evaluations of the microbiome in premature infants have shown decreased diversity of species and increased proportions of potentially pathogenic bacteria. Microbial patterns in premature infants may be affected by nutritional fat intake, altering risk of diseases such as necrotizing enterocolitis. Dietary FA may also impact disease susceptibility through molecular mechanisms. Specifically, intestinal Toll-like receptor 4 expression is altered by manipulation of FA in murine models. Abnormal increased expression of Toll-like receptor 4, the receptor for lipopolysaccharide, has been implicated in necrotizing enterocolitis. This report will review the role of dietary fat in the composition of the intestinal microbiome, the extreme variability of FA intake in premature infants, and associations of both dysbiosis and FA intake with the development of necrotizing enterocolitis.
Collapse
|
35
|
Beyond building better brains: bridging the docosahexaenoic acid (DHA) gap of prematurity. J Perinatol 2015; 35:1-7. [PMID: 25357095 PMCID: PMC4281288 DOI: 10.1038/jp.2014.195] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 08/23/2014] [Accepted: 08/26/2014] [Indexed: 12/12/2022]
Abstract
Long-chain polyunsaturated fatty acids (LCPUFA) including docosahexaenoic acid (DHA) are essential for normal vision and neurodevelopment. DHA accretion in utero occurs primarily in the last trimester of pregnancy to support rapid growth and brain development. Premature infants, born before this process is complete, are relatively deficient in this essential fatty acid. Very low birth weight (VLBW) infants remain deficient for a long period of time due to ineffective conversion from precursor fatty acids, lower fat stores and a limited nutritional provision of DHA after birth. In addition to long-term visual and neurodevelopmental risks, VLBW infants have significant morbidity and mortality from diseases specific to premature birth, including bronchopulmonary dysplasia, necrotizing enterocolitis, and retinopathy of prematurity. There is increasing evidence that DHA has protective benefits against these disease states. The aim of this article is to identify the unique needs of premature infants, review the current recommendations for LCPUFA provision in infants and discuss the caveats and innovative new ways to overcome the DHA deficiency through postnatal supplementation, with the long-term goal of improving morbidity and mortality in this at-risk population.
Collapse
|
36
|
Hsiao CC, Tsai ML, Chen CC, Lin HC. Early optimal nutrition improves neurodevelopmental outcomes for very preterm infants. Nutr Rev 2014; 72:532-40. [PMID: 24938866 DOI: 10.1111/nure.12110] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Chien-Chou Hsiao
- Department of Neonatology; Changhua Christian Hospital; Changhua Taiwan
- School of Medicine; Chung Shan Medical University; Taichung Taiwan
| | - Ming-Luen Tsai
- Department of Pediatrics; Children's Hospital; China Medical University; Taichung Taiwan
| | - Chih-Chen Chen
- Section of Neonatology; Department of Pediatrics; Kaohsiung Chang-Gung Memorial Hospital; Kaohsiung Taiwan
- College of Medicine; Chang-Gung University; Kaohsiung Taiwan
| | - Hung-Chih Lin
- Department of Pediatrics; Children's Hospital; China Medical University; Taichung Taiwan
- School of Chinese Medicine; China Medical University; Taichung Taiwan
| |
Collapse
|
37
|
Das U. HLA-DR expression, cytokines and bioactive lipids in sepsis. Arch Med Sci 2014; 10:325-35. [PMID: 24904669 PMCID: PMC4042054 DOI: 10.5114/aoms.2014.42586] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 01/03/2014] [Accepted: 01/04/2014] [Indexed: 12/17/2022] Open
Abstract
Sepsis accounts for more than 200,000 deaths annually in the USA alone. Both inflammatory and anti-inflammatory responses occur simultaneously in sepsis, the early phase dominated by the hyperinflammatory response and the late phase by immunosuppression. This late immunosuppression phase leads to loss of the delayed type hypersensitivity response, failure to clear the primary infection and development of secondary infections. Based on the available data, I hypothesize that failure to produce adequate amounts of inflammation resolving lipid mediators may be at the centre of both the hyperinflammatory response and late immunosuppression seen in sepsis. These proresolving lipids - lipoxins, resolvins and protectins - suppress exacerbated activation of leukocytes and macrophages, inhibit excess production of pro-inflammatory cytokines, initiate resolution of inappropriate inflammation, augment clearance of bacteria and other pathogens, and restore homeostasis. If true, this implies that administration of naturally occurring lipoxins, resolvins, protectins, maresins and nitrolipids by themselves or their more stable synthetic analogues such as 15-epi-16-(para-fluorophenoxy)-lipoxin A4-methyl ester, a synthetic analogue of 15-epi-lipoxin A4, and 15(R/S)-methyl-LXA4 may form a new approach in the prevention (in the high-risk subjects), management of sepsis and in resolving the imbalanced inflammatory process such that sepsis is ameliorated early. In addition, recent studies have suggested that nociceptin and cold inducible RNA binding protein (CIRBP) also have a role in the pathobiology of sepsis. It is suggested that both nociceptin and CIRBP inhibit the production of lipoxins, resolvins, protectins, maresins, and nitrolipids and thus play a role in sepsis and septic shock.
Collapse
|
38
|
Vegge A, Thymann T, Lauritzen L, Bering SB, Wiinberg B, Sangild PT. Parenteral lipids and partial enteral nutrition affect hepatic lipid composition but have limited short term effects on formula-induced necrotizing enterocolitis in preterm piglets. Clin Nutr 2014; 34:219-28. [PMID: 24703629 DOI: 10.1016/j.clnu.2014.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 03/07/2014] [Accepted: 03/08/2014] [Indexed: 11/18/2022]
Abstract
BACKGROUND & AIMS Rapid transition from total parenteral nutrition (TPN) to enteral feeding is a risk factor for necrotizing enterocolitis (NEC) in preterm infants. We hypothesized that partial enteral nutrition with colostrum, increased proportion of n-3 polyunsaturated fatty acids (PUFA), or exclusion of lipid in TPN would affect short term NEC sensitivity and liver function. METHODS Preterm piglets were fed for three days after birth: 1) TPN with a standard lipid emulsion (Nutriflex Lipid Plus, TPN control group, n = 19), 2) PN plus bovine colostrum as partial enteral nutrition (PN/COL, n = 18), 3) TPN with fish oil (FO) lipids (Omegaven, TPN/FO, n = 19), or 4) TPN with no lipid (TPN/NL, n = 22). After TPN, piglets were fed formula for two days before tissue collection. RESULTS None of the treatments had consistent effect on NEC incidence (∼40-50% across all groups), intestinal morphology and function, relative to TPN. In the liver, there were no signs of steatosis but PN/COL decreased the n-6 PUFA levels, leading to higher n-3/n-6 ratio, GGT activity, and plasma cholesterol and albumin levels, relative to TPN (all p < 0.05). TPN/FO increased the hepatic n-3 levels and n-3/n-6 ratio. TPN/NL treatment led to decreased hepatic n-6 level, n-3/n-6 ratio and bilirubin, albumin and triglycerides, and lowered blood clotting strength (-30%, TPN/NL vs. TPN/COL, p < 0.05). CONCLUSION Partial enteral nutrition with colostrum, increased n-3 PUFAs in TPN, or removal of lipid from the TPN, all affect hepatic lipids and proteins in preterm neonates. These effects do not translate into improved hepatic function or NEC resistance, at least not short term.
Collapse
Affiliation(s)
- Andreas Vegge
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark; Diabetes Pharmacology, Novo Nordisk A/S, Denmark
| | - Thomas Thymann
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Lotte Lauritzen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Stine B Bering
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Bo Wiinberg
- Department of Veterinary Clinical and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Haemophilia Pharmacology, Novo Nordisk A/S, Denmark
| | - Per T Sangild
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark.
| |
Collapse
|
39
|
Mustafi D, Shiou SR, Fan X, Markiewicz E, Karczmar GS, Claud EC. MRI of neonatal necrotizing enterocolitis in a rodent model. NMR IN BIOMEDICINE 2014; 27:272-279. [PMID: 24318809 PMCID: PMC4282586 DOI: 10.1002/nbm.3060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 11/11/2013] [Accepted: 11/12/2013] [Indexed: 06/02/2023]
Abstract
Neonatal necrotizing enterocolitis (NEC) is a poorly understood life-threatening illness afflicting premature infants. Research is hampered by the absence of a suitable method to monitor disease progression noninvasively. The primary goal of this research was to test in vivo MRI methods for the noninvasive early detection and staging of inflammation in the ileum of an infant rat model of NEC. Neonatal rats were delivered by cesarean section at embryonic stage of day 20 after the beginning of pregnancy and stressed with formula feeding, hypoxia and bacterial colonization to induce NEC. Naturally born and dam-fed neonatal rats were used as healthy controls. In vivo MRI studies were performed using a Bruker 9.4-T scanner to obtain high-resolution anatomical MR images using both gradient echo and spin echo sequences, pixel-by-pixel T2 maps using a multi-slice-multi-echo sequence, and maps of the apparent diffusion coefficient (ADC) of water using a spin echo sequence, to assess the degree of ileal damage. Pups were sacrificed at the end of the MRI experiment on day 2 or 4 for histology. T2 measured by MRI was increased significantly in the ileal regions of pups with NEC by histology (106.3 ± 6.1 ms) compared with experimentally stressed pups without NEC (85.2 ± 6.8 ms) and nonstressed, control rat pups (64.9 ± 2.3 ms). ADC values measured by diffusion-weighted MRI were also increased in the ileal regions of pups with NEC by histology [(1.98 ± 0.15) × 10(-3) mm(2)/s] compared with experimentally stressed pups without NEC [(1.43 ± 0.16) × 10(-3) mm(2)/s] and nonstressed control pups [(1.10 ± 0.06) × 10(-3) mm(2)/s]. Both T2 and ADC values between these groups were found to be significantly different (p < 0.03). The correlation of MRI results with histologic images of the excised ileal tissue samples strongly suggests that MRI can noninvasively identify NEC and assess intestinal injury prior to clinical symptoms in a physiologic rat pup model of NEC.
Collapse
Affiliation(s)
| | - Sheng-Ru Shiou
- Department of Pediatrics, University of ChicagoChicago, IL, USA
| | - Xiaobing Fan
- Department of Radiology, University of ChicagoChicago, IL, USA
| | | | | | - Erika C Claud
- Department of Pediatrics, University of ChicagoChicago, IL, USA
- Department of Medicine, University of ChicagoChicago, IL, USA
| |
Collapse
|
40
|
Xiao G, Tang L, Yuan F, Zhu W, Zhang S, Liu Z, Geng Y, Qiu X, Zhang Y, Su L. Eicosapentaenoic acid enhances heat stress-impaired intestinal epithelial barrier function in Caco-2 cells. PLoS One 2013; 8:e73571. [PMID: 24066055 PMCID: PMC3774713 DOI: 10.1371/journal.pone.0073571] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Accepted: 07/27/2013] [Indexed: 02/03/2023] Open
Abstract
Objective Dysfunction of the intestinal epithelial tight junction (TJ) barrier is known to have an important etiologic role in the pathophysiology of heat stroke. N-3 polyunsaturated fatty acids (PUFAs), including eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), play a role in maintaining and protecting the TJ structure and function. This study is aimed at investigating whether n-3 PUFAs could alleviate heat stress-induced dysfunction of intestinal tight junction. Methods Human intestinal epithelial Caco-2 cells were pre-incubated with EPA, DHA or arachidonic acid (AA) and then exposed to heat stress. Transepithelial electrical resistance (TEER) and Horseradish Peroxidase (HRP) permeability were measured to analyze barrier integrity. Levels of TJ proteins, including occludin, ZO-1 and claudin-2, were analyzed by Western blot and localized by immunofluorescence microscopy. Messenger RNA levels were determined by quantitative real time polymerase chain reaction (Q-PCR). TJ morphology was observed by transmission electron microscopy. Results EPA effectively attenuated the decrease in TEER and impairment of intestinal permeability in HRP flux induced by heat exposure. EPA significantly elevated the expression of occludin and ZO-1, while DHA was less effective and AA was not at all effective. The distortion and redistribution of TJ proteins, and disruption of morphology were also effectively prevented by pretreatment with EPA. Conclusion This study indicates for the first time that EPA is more potent than DHA in protecting against heat-induced permeability dysfunction and epithelial barrier damage of tight junction.
Collapse
Affiliation(s)
- Guizhen Xiao
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liqun Tang
- Key Laboratory of Hot Zone Trauma Care and Tissue Repair of PLA, Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Fangfang Yuan
- Key Laboratory of Hot Zone Trauma Care and Tissue Repair of PLA, Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Wei Zhu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaoheng Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhifeng Liu
- Key Laboratory of Hot Zone Trauma Care and Tissue Repair of PLA, Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Yan Geng
- Key Laboratory of Hot Zone Trauma Care and Tissue Repair of PLA, Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Xiaowen Qiu
- Department of Nutrition, General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Yali Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- * E-mail: (YZ); (LS)
| | - Lei Su
- Key Laboratory of Hot Zone Trauma Care and Tissue Repair of PLA, Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou, China
- * E-mail: (YZ); (LS)
| |
Collapse
|
41
|
Influence of genotype on the modulation of gene and protein expression by n-3 LC-PUFA in rats. GENES AND NUTRITION 2013; 8:589-600. [PMID: 23744008 DOI: 10.1007/s12263-013-0349-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 05/22/2013] [Indexed: 01/22/2023]
Abstract
It is becoming increasingly apparent that responsiveness to dietary fat composition is heterogeneous and dependent on the genetic make-up of the individual. The aim of this study was to evidence a genotype-related differential effect of n-3 long-chain polyunsaturated fatty acids (n-3 LC-PUFA) on the modulation of hepatic genes involved in cholesterol metabolism. Fourteen spontaneously hypertensive (SH) rats, which present a naturally occurring variation in the gene encoding for sterol responsive element binding protein 1 (SREBP-1), contributing to their inherited variation in lipid metabolism, and 14 Wistar-Kyoto (WK) rats were fed a control diet or an n-3 LC-PUFA enriched diet for 90 days. Plasma lipid profile, total lipid fatty acid composition in plasma and liver, and the expression of SREBP-1 and 2, 3-hydroxy-3-methyl-glutaryl-CoA reductase, low-density lipoprotein receptor, and acyl-CoA:cholesterol acyltransferase 2 encoding genes and proteins were determined. The positive effect of the enriched diet on the serum lipid profile, particularly on total cholesterol and triglyceride level, was clearly evidenced in both WK and SH rats, but n-3 LC-PUFA acted through a different modulation of gene and protein expression that appeared related to the genetic background. Our study evidences a different transcriptional effect of specific nutrients related to genetic variants.
Collapse
|
42
|
Is sepsis a pro-resolution deficiency disorder? Med Hypotheses 2013; 80:297-9. [DOI: 10.1016/j.mehy.2012.12.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 12/05/2012] [Indexed: 12/19/2022]
|
43
|
Coursodon-Boyiddle CF, Snarrenberg CL, Adkins-Rieck CK, Bassaganya-Riera J, Hontecillas R, Lawrence P, Brenna JT, Jouni ZE, Dvorak B. Pomegranate seed oil reduces intestinal damage in a rat model of necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol 2012; 303:G744-51. [PMID: 22821948 PMCID: PMC3468537 DOI: 10.1152/ajpgi.00248.2012] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Pomegranate seed oil (PSO), which is the major source of conjugated linolenic acids such as punicic acid (PuA), exhibits strong anti-inflammatory properties. Necrotizing enterocolitis (NEC) is a devastating disease associated with severe and excessive intestinal inflammation. The aim of this study was to evaluate the effects of orally administered PSO on the development of NEC, intestinal epithelial proliferation, and cytokine regulation in a rat model of NEC. Premature rats were divided into three groups: dam fed (DF), formula-fed rats (FF), or rats fed with formula supplemented with 1.5% of PSO (FF + PSO). All groups were exposed to asphyxia/cold stress to induce NEC. Intestinal injury, epithelial cell proliferation, cytokine production, and trefoil factor 3 (Tff3) production were evaluated in the terminal ileum. Oral administration of PSO (FF+PSO) decreased the incidence of NEC from 61 to 26%. Feeding formula with PSO improved enterocyte proliferation in the site of injury. Increased levels of proinflammatory IL-6, IL-8, IL-12, IL-23, and TNF-α in the ileum of FF rats were normalized in PSO-treated animals. Tff3 production in the FF rats was reduced compared with DF but not further affected by the PSO. In conclusion, administration of PSO protects against NEC in the neonatal rat model. This protective effect is associated with an improvement of intestinal epithelial homeostasis and a strong anti-inflammatory effect of PSO on the developing intestinal mucosa.
Collapse
Affiliation(s)
| | - Chelsea L. Snarrenberg
- 1Department of Pediatrics and Steele Children's Research Center, The University of Arizona, Tucson, Arizona;
| | - Camille K. Adkins-Rieck
- 1Department of Pediatrics and Steele Children's Research Center, The University of Arizona, Tucson, Arizona;
| | - Josep Bassaganya-Riera
- 2Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia;
| | - Raquel Hontecillas
- 2Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia;
| | - Peter Lawrence
- 3Division of Nutritional Sciences, Cornell University, Ithaca, New York;
| | - J. Thomas Brenna
- 3Division of Nutritional Sciences, Cornell University, Ithaca, New York;
| | | | - Bohuslav Dvorak
- 1Department of Pediatrics and Steele Children's Research Center, The University of Arizona, Tucson, Arizona;
| |
Collapse
|
44
|
Valentine CJ. Maternal dietary DHA supplementation to improve inflammatory outcomes in the preterm infant. Adv Nutr 2012; 3:370-6. [PMID: 22585914 PMCID: PMC3649472 DOI: 10.3945/an.111.001248] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Dietary DHA (22:6n-3) is a long-chain PUFA that has provocative effects on inflammatory signal events that could potentially affect preterm infant health. It is well known that the essential fatty acid of the (n-3) series; α-linolenic acid (18:3n:3) can be desaturated and elongated in the liver endoplasmic reticulum and peroxisome to produce the 22-carbon DHA. Nevertheless, concern exists as to the efficiency of this mechanism in providing the preterm infant with adequate DHA. Activity of the δ-6-desaturase and the δ-5-desaturase necessary for DHA synthesis is decreased by protein deprivation. The combined effects of suboptimal intake of both DHA and protein in the preterm infants could have substantial clinical consequences.
Collapse
Affiliation(s)
- Christina J Valentine
- Division of Neonatology, Perinatal, and Pulmonary Biology, Center for Interdisciplinary Research in Human Milk and Lactation, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
45
|
Morales E, García-Esteban R, Guxens M, Guerra S, Mendez M, Moltó-Puigmartí C, Lopez-Sabater MC, Sunyer J. Effects of prolonged breastfeeding and colostrum fatty acids on allergic manifestations and infections in infancy. Clin Exp Allergy 2012; 42:918-28. [DOI: 10.1111/j.1365-2222.2012.03969.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- E. Morales
- Center for Research in Environmental Epidemiology (CREAL); Barcelona Catalonia Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública; Barcelona Catalonia Spain
| | - R. García-Esteban
- Center for Research in Environmental Epidemiology (CREAL); Barcelona Catalonia Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública; Barcelona Catalonia Spain
| | - M. Guxens
- Center for Research in Environmental Epidemiology (CREAL); Barcelona Catalonia Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública; Barcelona Catalonia Spain
| | - S. Guerra
- Center for Research in Environmental Epidemiology (CREAL); Barcelona Catalonia Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública; Barcelona Catalonia Spain
- Arizona Respiratory Center; Tucson AZ USA
| | - M. Mendez
- Center for Research in Environmental Epidemiology (CREAL); Barcelona Catalonia Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública; Barcelona Catalonia Spain
| | - C. Moltó-Puigmartí
- CIBER Epidemiología y Salud Pública; Barcelona Catalonia Spain
- Department of Nutrition and Food Science; Faculty of Pharmacy; University of Barcelona; Barcelona Catalonia Spain
| | - M. C. Lopez-Sabater
- CIBER Epidemiología y Salud Pública; Barcelona Catalonia Spain
- Department of Nutrition and Food Science; Faculty of Pharmacy; University of Barcelona; Barcelona Catalonia Spain
| | - J. Sunyer
- Center for Research in Environmental Epidemiology (CREAL); Barcelona Catalonia Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública; Barcelona Catalonia Spain
- Department of Experimental and Health Sciences; Pompeu Fabra University; Barcelona Catalonia Spain
| |
Collapse
|
46
|
Ran-Ressler RR, Khailova L, Arganbright KM, Adkins-Rieck CK, Jouni ZE, Koren O, Ley RE, Brenna JT, Dvorak B. Branched chain fatty acids reduce the incidence of necrotizing enterocolitis and alter gastrointestinal microbial ecology in a neonatal rat model. PLoS One 2011; 6:e29032. [PMID: 22194981 PMCID: PMC3237582 DOI: 10.1371/journal.pone.0029032] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 11/19/2011] [Indexed: 01/05/2023] Open
Abstract
Introduction Branched chain fatty acids (BCFA) are found in the normal term human newborn's gut, deposited as major components of vernix caseosa ingested during late fetal life. We tested the hypothesis that premature infants' lack of exposure to gastrointestinal (GI) BCFA is associated with their microbiota and risk for necrotizing enterocolitis (NEC) using a neonatal rat model. Methods Pups were collected one day before scheduled birth. The pups were exposed to asphyxia and cold stress to induce NEC. Pups were assigned to one of three experimental treatments. DF (dam-fed) ; Control, hand-fed rat milk substitute ; BCFA, hand-fed rat milk substitute with 20%w/w BCFA. Total fat was equivalent (11%wt) for both the Control and BCFA groups. Cecal microbiota were characterized by 16S rRNA gene pyrosequencing, and intestinal injury, ileal cytokine and mucin gene expression, interleukin-10 (IL-10) peptide immunohistochemistry, and BCFA uptake in ileum phospholipids, serum and liver were assessed. Results NEC incidence was reduced by over 50% in the BCFA group compared to the Control group as assessed in ileal tissue; microbiota differed among all groups. BCFA-fed pups harbored greater levels of BCFA-associated Bacillus subtilis and Pseudomonas aeruginosa compared to Controls. Bacillus subtilis levels were five-fold greater in healthy pups compared to pups with NEC. BCFA were selectively incorporated into ileal phospholipids, serum and liver tissue. IL-10 expression increased three-fold in the BCFA group versus Controls and no other inflammatory or mucosal mRNA markers changed. Conclusion At constant dietary fat level, BCFA reduce NEC incidence and alter microbiota composition. BCFA are also incorporated into pup ileum where they are associated with enhanced IL-10 and may exert other specific effects.
Collapse
Affiliation(s)
- Rinat R. Ran-Ressler
- Division of Nutritional Sciences, Savage Hall, Cornell University, Ithaca, New York, United States of America
| | - Ludmila Khailova
- Department of Pediatrics, University of Arizona, Tucson, Arizona, United States of America
| | - Kelly M. Arganbright
- Department of Pediatrics, University of Arizona, Tucson, Arizona, United States of America
| | | | - Zeina E. Jouni
- Mead Johnson Nutrition, Evansville, Indiana, United States of America
| | - Omry Koren
- Department of Microbiology, Cornell University, Ithaca, New York, United States of America
| | - Ruth E. Ley
- Department of Microbiology, Cornell University, Ithaca, New York, United States of America
| | - J. Thomas Brenna
- Division of Nutritional Sciences, Savage Hall, Cornell University, Ithaca, New York, United States of America
- * E-mail: (JTB); (BD)
| | - Bohuslav Dvorak
- Department of Pediatrics, University of Arizona, Tucson, Arizona, United States of America
- * E-mail: (JTB); (BD)
| |
Collapse
|
47
|
Siggers RH, Siggers J, Thymann T, Boye M, Sangild PT. Nutritional modulation of the gut microbiota and immune system in preterm neonates susceptible to necrotizing enterocolitis. J Nutr Biochem 2010; 22:511-21. [PMID: 21193301 DOI: 10.1016/j.jnutbio.2010.08.002] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2010] [Accepted: 08/23/2010] [Indexed: 02/07/2023]
Abstract
The gastrointestinal inflammatory disorder, necrotizing enterocolitis (NEC), is among the most serious diseases for preterm neonates. Nutritional, microbiological and immunological dysfunctions all play a role in disease progression but the relationship among these determinants is not understood. The preterm gut is very sensitive to enteral feeding which may either promote gut adaptation and health, or induce gut dysfunction, bacterial overgrowth and inflammation. Uncontrolled inflammatory reactions may be initiated by maldigestion and impaired mucosal protection, leading to bacterial overgrowth and excessive nutrient fermentation. Tumor necrosis factor alpha, toll-like receptors and heat-shock proteins are identified among the immunological components of the early mucosal dysfunction. It remains difficult, however, to distinguish the early initiators of NEC from the later consequences of the disease pathology. To elucidate the mechanisms and identify clinical interventions, animal models showing spontaneous NEC development after preterm birth coupled with different forms of feeding may help. In this review, we summarize the literature and some recent results from studies on preterm pigs on the nutritional, microbial and immunological interactions during the early feeding-induced mucosal dysfunction and later NEC development. We show that introduction of suboptimal enteral formula diets, coupled with parenteral nutrition, predispose to disease, while advancing amounts of mother's milk from birth (particularly colostrum) protects against disease. Hence, the transition from parenteral to enteral nutrition shortly after birth plays a pivotal role to secure gut growth, digestive maturation and an appropriate response to bacterial colonization in the sensitive gut of preterm neonates.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Enterocolitis, Necrotizing/etiology
- Enterocolitis, Necrotizing/immunology
- Enterocolitis, Necrotizing/microbiology
- Gastrointestinal Tract/growth & development
- Gastrointestinal Tract/immunology
- Gastrointestinal Tract/microbiology
- Heat-Shock Proteins/metabolism
- Humans
- Immune System/immunology
- Infant Nutritional Physiological Phenomena
- Infant, Newborn
- Infant, Premature
- Infant, Premature, Diseases/etiology
- Infant, Premature, Diseases/immunology
- Infant, Premature, Diseases/microbiology
- Intestinal Mucosa/immunology
- Intestinal Mucosa/microbiology
- Intestine, Small/metabolism
- Metagenome/physiology
Collapse
Affiliation(s)
- Richard H Siggers
- Department of Human Nutrition, Faculty of Life Sciences, University of Copenhagen, 30 Rolighedsvej, DK-1958 Frederiksberg C, Denmark
| | | | | | | | | |
Collapse
|
48
|
Josephson CD, Wesolowski A, Bao G, Sola-Visner MC, Dudell G, Castillejo MI, Shaz BH, Easley KA, Hillyer CD, Maheshwari A. Do red cell transfusions increase the risk of necrotizing enterocolitis in premature infants? J Pediatr 2010; 157:972-978.e1-3. [PMID: 20650470 PMCID: PMC4425198 DOI: 10.1016/j.jpeds.2010.05.054] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 04/23/2010] [Accepted: 05/28/2010] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To test the hypothesis that red blood cell (RBC) transfusions increase the risk of necrotizing enterocolitis (NEC) in premature infants, we investigated whether the risk of "transfusion-associated" NEC is higher in infants with lower hematocrits and advanced postnatal age. STUDY DESIGN Retrospective comparison of NEC patients and control patients born at < 34 weeks gestation. RESULTS The frequency of RBC transfusions was similar in NEC patients (47/93, 51%) and control patients (52/91, 58%). Late-onset NEC (> 4 weeks of age) was more frequently associated with a history of transfusion(s) than early-onset NEC (adjusted OR, 6.7; 95% CI, 1.5 to 31.2; P = .02). Compared with nontransfused patients, RBC-transfused patients were born at earlier gestational ages, had greater intensive care needs (including at the time of onset of NEC), and longer hospital stay. A history of RBC transfusions within 48-hours before NEC onset was noted in 38% of patients, most of whom were extremely low birth weight infants. CONCLUSIONS In most patients, RBC transfusions were temporally unrelated to NEC and may be merely a marker of overall severity of illness. However, the relationship between RBC transfusions and NEC requires further evaluation in extremely low birth weight infants using a prospective cohort design.
Collapse
Affiliation(s)
- Cassandra D. Josephson
- Department of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Emory University School of Medicine, Atlanta, GA
- Aflac Cancer Center & Blood Disorders Services at Children’s Healthcare of Atlanta, Atlanta, GA
| | | | - Gaobin Bao
- Biostatistics and Bioinformatics, Rollins School of Public Health, Atlanta, GA
| | - Martha C. Sola-Visner
- Department of Pediatrics, Division of Newborn Medicine, Children’s Hospital and Harvard Medical School, Boston, MA
| | - Golde Dudell
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Children’s Hospital and Research Center, Oakland, CA
| | - Marta-Inés Castillejo
- Department of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Emory University School of Medicine, Atlanta, GA
| | - Beth H. Shaz
- Department of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Emory University School of Medicine, Atlanta, GA
| | - Kirk A. Easley
- Biostatistics and Bioinformatics, Rollins School of Public Health, Atlanta, GA
| | | | - Akhil Maheshwari
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
49
|
Brignardello J, Morales P, Diaz E, Romero J, Brunser O, Gotteland M. Pilot study: alterations of intestinal microbiota in obese humans are not associated with colonic inflammation or disturbances of barrier function. Aliment Pharmacol Ther 2010; 32:1307-14. [PMID: 21050232 DOI: 10.1111/j.1365-2036.2010.04475.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Obesity is associated with low-grade inflammation contributing to insulin-resistance. Gut barrier alterations, described in animal models of obesity, probably favour inflammation. This has not been hitherto described in obese humans. AIM To evaluate gut permeability in asymptomatic obese and its association with plasma (C-reactive protein (CRP), arachidonate/eicosapentaenoate ratio) and faecal (calprotectin and leptin) markers of inflammation and microbiota alterations. METHODS A total of 13 obese (age: 33.9 ± 11.5 years; BMI: 35.9 ± 5.0 kg/m²) and 11 control subjects (age: 30.3 ± 8.1 years; BMI: 23.5 ± 2.4 kg/m²) were recruited. Gut permeability was assessed by the lactulose-mannitol-sucralose test, plasma fatty acids by gas chromatography, faecal calprotectin and leptin by Elisa and faecal microbiota by G+C profiling. RESULTS C-reactive protein was increased in the obese subjects (P = 0.01), but neither the plasma arachidonate/eicosapentaenoate ratio, the faecal levels of calprotectin and leptin, nor the gut permeability were altered. The faecal microbiota was altered in the obese (P = 0.0002), with predominance of bacterial populations having a lower G+C content and decreased concentrations of high G+C populations. CONCLUSIONS Asymptomatic obese individuals with systemic low-grade inflammation do not have evidence of colonic inflammation or gut barrier alteration; however, the biodiversity of their intestinal microbiota is affected.
Collapse
Affiliation(s)
- J Brignardello
- Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | | | | | | | | | | |
Collapse
|
50
|
Brilli RJ, McClead RE, Davis T, Stoverock L, Rayburn A, Berry JC, Shaz BH, Easley KA, Hillyer CD, Maheshwari A. The Preventable Harm Index: an effective motivator to facilitate the drive to zero. J Pediatr 2010; 157:681-3. [PMID: 20650469 DOI: 10.1016/j.jpeds.2010.05.046] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Revised: 03/31/2010] [Accepted: 05/26/2010] [Indexed: 11/26/2022]
Affiliation(s)
- Richard J Brilli
- Nationwide Children's Hospital, Department of Pediatrics, Division of Critical Care Medicine, The Ohio State University College of Medicine, Columbus, OH 43205, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|