1
|
Agakidou E, Agakidis C, Gika H, Sarafidis K. Emerging Biomarkers for Prediction and Early Diagnosis of Necrotizing Enterocolitis in the Era of Metabolomics and Proteomics. Front Pediatr 2020; 8:602255. [PMID: 33425815 PMCID: PMC7793899 DOI: 10.3389/fped.2020.602255] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022] Open
Abstract
Necrotizing Enterocolitis (NEC) is a catastrophic disease affecting predominantly premature infants and is characterized by high mortality and serious long-term consequences. Traditionally, diagnosis of NEC is based on clinical and radiological findings, which, however, are non-specific for NEC, thus confusing differential diagnosis of other conditions such as neonatal sepsis and spontaneous intestinal perforation. In addition, by the time clinical and radiological findings become apparent, NEC has already progressed to an advanced stage. During the last three decades, a lot of research has focused on the discovery of biomarkers, which could accurately predict and make an early diagnosis of NEC. Biomarkers used thus far in clinical practice include acute phase proteins, inflammation mediators, and molecules involved in the immune response. However, none has been proven accurate enough to predict and make an early diagnosis of NEC or discriminate clinical from surgical NEC or other non-NEC gastrointestinal diseases. Complexity of mechanisms involved in NEC pathogenesis, which remains largely poorly elucidated, could partly explain the unsatisfactory diagnostic performance of the existing NEC biomarkers. More recently applied technics can provide important insight into the pathophysiological mechanisms underlying NEC but can also aid the detection of potentially predictive, early diagnostic, and prognostic biomarkers. Progress in omics technology has allowed for the simultaneous measurement of a large number of proteins, metabolic products, lipids, and genes, using serum/plasma, urine, feces, tissues, and other biological specimens. This review is an update of current data on emerging NEC biomarkers detected using proteomics and metabolomics, further discussing limitations and future perspectives in prediction and early diagnosis of NEC.
Collapse
Affiliation(s)
- Eleni Agakidou
- 1st Department of Neonatology, Faculty of Medicine, Ippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Charalampos Agakidis
- 1st Department of Pediatrics, Faculty of Medicine, Ippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Helen Gika
- Laboratory of Forensic Medicine and Toxicology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece.,BIOMIC_AUTH, Bioanalysis and Omics Laboratory, Centre for Interdisciplinary Research and Innovation, CIRI-AUTH B1.4, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kosmas Sarafidis
- 1st Department of Neonatology, Faculty of Medicine, Ippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
2
|
Piglet birth weight, subsequent performance, carcass traits and pork quality: A meta-analytical study. Livest Sci 2018. [DOI: 10.1016/j.livsci.2018.05.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
3
|
Jiang P, Vegge A, Thymann T, Wan JMF, Sangild PT. Glucagon-Like Peptide 2 Stimulates Postresection Intestinal Adaptation in Preterm Pigs by Affecting Proteins Related to Protein, Carbohydrate, and Sulphur Metabolism. JPEN J Parenter Enteral Nutr 2016; 41:1293-1300. [DOI: 10.1177/0148607116662971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Pingping Jiang
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Andreas Vegge
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
- Global Research, Novo Nordisk, Måløv, Denmark
| | - Thomas Thymann
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Per Torp Sangild
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
4
|
Chen F, Wang T, Feng C, Lin G, Zhu Y, Wu G, Johnson G, Wang J. Proteome Differences in Placenta and Endometrium between Normal and Intrauterine Growth Restricted Pig Fetuses. PLoS One 2015; 10:e0142396. [PMID: 26554841 PMCID: PMC4640832 DOI: 10.1371/journal.pone.0142396] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 10/21/2015] [Indexed: 01/21/2023] Open
Abstract
Uteroplacental tissue plays a key role in substance exchanges between maternal and fetal circulation, and, therefore, in the growth and development of fetuses. In this study, proteomics and western blotting were applied to investigate the changes of proteome in the placenta and endometrium of normal and intrauterine growth restriction (IUGR) porcine fetuses during mid to late pregnancy (D60, 90, and 110 of gestation). Our results showed that proteins participating in cell structure, energy metabolism, stress response, cell turnover, as well as transport and metabolism of nutrients were differentially expressed in placenta and endometrium between normal and IUGR fetuses. Analysis of functions of these proteins suggests reductions in ATP production and nutrients transport, increases in oxidative stress and apoptosis, and impairment of cell metabolism in IUGR fetuses. Collectively, our findings aid in understanding of the mechanisms responsible for uteroplacental dysfunction in IUGR fetus, and are expected to provide new strategies to reduce fetal growth restriction in pigs and other mammals.
Collapse
Affiliation(s)
- Fang Chen
- State Key Laboratory of Animal Nutrition, Alltech-MAFIC Research Alliance, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Department of Animal Science, South China Agricultural University, Guangzhou, China
| | - Taiji Wang
- State Key Laboratory of Animal Nutrition, Alltech-MAFIC Research Alliance, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Cuiping Feng
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, China
| | - Gang Lin
- State Key Laboratory of Animal Nutrition, Alltech-MAFIC Research Alliance, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yuhua Zhu
- State Key Laboratory of Animal Nutrition, Alltech-MAFIC Research Alliance, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Guoyao Wu
- State Key Laboratory of Animal Nutrition, Alltech-MAFIC Research Alliance, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Department of Animal Science, Texas A&M University, College Station, Texas, United States of America
| | - Gregory Johnson
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, United States of America
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, Alltech-MAFIC Research Alliance, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
5
|
Bassols A, Costa C, Eckersall PD, Osada J, Sabrià J, Tibau J. The pig as an animal model for human pathologies: A proteomics perspective. Proteomics Clin Appl 2014; 8:715-31. [DOI: 10.1002/prca.201300099] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 04/28/2014] [Accepted: 07/30/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Anna Bassols
- Departament de Bioquímica i Biologia Molecular; Facultat de Veterinària; Universitat Autònoma de Barcelona; Cerdanyola del Vallès Spain
| | - Cristina Costa
- New Therapies of Genes and Transplants Group; Institut d'Investigació Biomèdica de Bellvitge (IDIBELL); L'Hospitalet de Llobregat; Barcelona Spain
| | - P. David Eckersall
- Institute of Biodiversity, Animal Health and Comparative Medicine; University of Glasgow; Glasgow UK
| | - Jesús Osada
- Departamento de Bioquímica y Biología Molecular; Facultad de Ciencias; Universidad de Zaragoza; CIBEROBN; Zaragoza Spain
| | - Josefa Sabrià
- Departament de Bioquímica i Biologia Molecular; Facultat de Medicina; Institut de Neurociències (INc); Universitat Autònoma de Barcelona; Cerdanyola del Vallès Spain
| | - Joan Tibau
- IRTA - Food Technology; Animal Genetics Program; Finca Camps i Armet; Monells Spain
| |
Collapse
|
6
|
Liu Y, Tran DQ, Fatheree NY, Marc Rhoads J. Lactobacillus reuteri DSM 17938 differentially modulates effector memory T cells and Foxp3+ regulatory T cells in a mouse model of necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol 2014; 307:G177-86. [PMID: 24852566 PMCID: PMC4101683 DOI: 10.1152/ajpgi.00038.2014] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Necrotizing enterocolitis (NEC) is an inflammatory disease with evidence of increased production of proinflammatory cytokines in the intestinal mucosa. Lactobacillus reuteri DSM 17938 (LR17938) has been shown to have anti-inflammatory activities in an experimental model of NEC. Activated effector lymphocyte recruitment to sites of inflammation requires the sequential engagement of adhesion molecules such as CD44. The phenotype of CD44(+)CD45RB(lo) separates T effector/memory (Tem) cells from naive (CD44(-)CD45RB(hi)) cells. It is unknown whether these Tem cells participate in the inflammation associated with NEC and can be altered by LR17938. NEC was induced in 8- to 10-day-old C57BL/6J mice by gavage feeding with formula and exposure to hypoxia and cold stress for 4 days. Survival curves and histological scores were analyzed. Lymphocytes isolated from mesenteric lymph nodes and ileum were labeled for CD4, CD44, CD45RB, intracellular Foxp3, and Helios and subsequently analyzed by flow cytometry. LR17938 decreased mortality and the incidence and severity of NEC. The percentage of Tem cells in the ileum and mesenteric lymph nodes was increased in NEC but decreased by LR17938. Conversely, the percentage of CD4(+)Foxp3(+) regulatory T (Treg) cells in the intestine decreased during NEC and was restored to normal by LR17938. The majority of the Treg cells preserved by LR17938 were Helios+ subsets, possibly of thymic origin. In conclusion, LR17938 may represent a useful treatment to prevent NEC. The mechanism of protection by LR17938 involves modulation of the balance between Tem and Treg cells. These T cell subsets might be potential biomarkers and therapeutic targets during intestinal inflammation.
Collapse
Affiliation(s)
- Yuying Liu
- 1Division of Gastroenterology, Department of Pediatrics, University of Texas Health Science Center at Houston Medical School, Houston, Texas; ,3Pediatric Research Center, University of Texas Health Science Center at Houston Medical School, Houston, Texas
| | - Dat Q. Tran
- 2Division of Allergy/Immunology/Rheumatology, Department of Pediatrics, University of Texas Health Science Center at Houston Medical School, Houston, Texas; and ,3Pediatric Research Center, University of Texas Health Science Center at Houston Medical School, Houston, Texas
| | - Nicole Y. Fatheree
- 1Division of Gastroenterology, Department of Pediatrics, University of Texas Health Science Center at Houston Medical School, Houston, Texas;
| | - J. Marc Rhoads
- 1Division of Gastroenterology, Department of Pediatrics, University of Texas Health Science Center at Houston Medical School, Houston, Texas; ,3Pediatric Research Center, University of Texas Health Science Center at Houston Medical School, Houston, Texas
| |
Collapse
|
7
|
Jiang P, Sangild PT. Intestinal proteomics in pig models of necrotising enterocolitis, short bowel syndrome and intrauterine growth restriction. Proteomics Clin Appl 2014; 8:700-14. [PMID: 24634357 DOI: 10.1002/prca.201300097] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 01/16/2014] [Accepted: 03/11/2014] [Indexed: 12/13/2022]
Abstract
Necrotising enterocolitis (NEC), short bowel syndrome (SBS) and intrauterine growth restriction (IUGR) are three conditions associated with intestinal dysfunction in newborn infants, particularly those born preterm. Piglet (Sus scrofa) models have recently been developed for NEC, SBS and IUGR, and tissue proteomic analyses have identified unknown pathways and new prognostic disease markers. Intestinal HSPs, iron metabolism proteins and proteins related to amino acid (e.g. arginine) and glucose metabolism are consistently affected by NEC progression and some of these proteins are also affected by SBS and IUGR. Parallel changes in some plasma and urinary proteins (e.g. haptoglobin, globulins, complement proteins, fatty acid binding proteins) may mirror the intestinal responses and pave the way to biomarker discovery. Explorative non-targeted proteomics provides ideas about the cellular pathways involved in intestinal adaptation during the critical neonatal period. Proteomics, combined with other -omic techniques, helps to get a more holistic picture of intestinal adaptation during NEC, SBS and IUGR. Explorative -omic research methods also have limitations and cannot replace, but only supplement, classical hypothesis-driven research that investigate disease mechanisms using a single or few endpoints.
Collapse
Affiliation(s)
- Pingping Jiang
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg, Denmark
| | | |
Collapse
|
8
|
Ceciliani F, Restelli L, Lecchi C. Proteomics in farm animals models of human diseases. Proteomics Clin Appl 2014; 8:677-88. [PMID: 24595991 DOI: 10.1002/prca.201300080] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 11/18/2013] [Accepted: 01/14/2014] [Indexed: 12/21/2022]
Abstract
The need to provide in vivo complex environments to understand human diseases strongly relies on the use of animal models, which traditionally include small rodents and rabbits. It is becoming increasingly evident that the few species utilised to date cannot be regarded as universal. There is a great need for new animal species that are naturally endowed with specific features relevant to human diseases. Farm animals, including pigs, cows, sheep and horses, represent a valid alternative to commonly utilised rodent models. There is an ample scope for the application of proteomic techniques in farm animals, and the establishment of several proteomic maps of plasma and tissue has clearly demonstrated that farm animals provide a disease environment that closely resembles that of human diseases. The present review offers a snapshot of how proteomic techniques have been applied to farm animals to improve their use as biomedical models. Focus will be on specific topics of biomedical research in which farm animal models have been characterised through the application of proteomic techniques.
Collapse
Affiliation(s)
- Fabrizio Ceciliani
- Department of Veterinary Sciences and Public Health, Università di Milano, Milan, Italy; Interdepartmental Centre for Studies on Mammary Gland, Università di Milano, Milan, Italy
| | | | | |
Collapse
|
9
|
Kandasamy J, Huda S, Ambalavanan N, Jilling T. Inflammatory signals that regulate intestinal epithelial renewal, differentiation, migration and cell death: Implications for necrotizing enterocolitis. ACTA ACUST UNITED AC 2014; 21:67-80. [PMID: 24533974 DOI: 10.1016/j.pathophys.2014.01.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Necrotizing enterocolitis is a disease entity with multiple proposed pathways of pathogenesis. Various combinations of these risk factors, perhaps based on genetic predisposition, possibly lead to the mucosal and epithelial injury that is the hallmark of NEC. Intestinal epithelial integrity is controlled by a tightly regulated balance between proliferation and differentiation of epithelium from intestinal epithelial stem cells and cellular loss by apoptosis. various signaling pathways play a key role in creating and maintaining this balance. The aim of this review article is to outline intestinal epithelial barrier development and structure and the impact of these inflammatory signaling and regulatory pathways as they pertain to the pathogenesis of NEC.
Collapse
Affiliation(s)
- Jegen Kandasamy
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Alabama at Birmingham, USA
| | - Shehzad Huda
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Alabama at Birmingham, USA
| | - Namasivayam Ambalavanan
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Alabama at Birmingham, USA
| | - Tamas Jilling
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Alabama at Birmingham, USA.
| |
Collapse
|
10
|
Sangild PT, Thymann T, Schmidt M, Stoll B, Burrin DG, Buddington RK. Invited review: the preterm pig as a model in pediatric gastroenterology. J Anim Sci 2013; 91:4713-29. [PMID: 23942716 DOI: 10.2527/jas.2013-6359] [Citation(s) in RCA: 196] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
At birth, the newborn mammal undergoes a transition from a sterile uterine environment with a constant nutrient supply, to a microbe-rich environment with intermittent oral intake of complex milk nutrients via the gastrointestinal tract (GIT). These functional challenges partly explain the relatively high morbidity and mortality of neonates. Preterm birth interrupts prenatal organ maturation, including that of the GIT, and increases disease risk. Exemplary is necrotizing enterocolitis (NEC), which is associated closely with GIT immaturity, enteral feeding, and bacterial colonization. Infants with NEC may require resection of the necrotic parts of the intestine, leading to short bowel syndrome (SBS), characterized by reduced digestive capacity, fluid loss, and dependency on parenteral nutrition. This review presents the preterm pig as a translational model in pediatric gastroenterology that has provided new insights into important pediatric diseases such as NEC and SBS. We describe protocols for delivery, care, and handling of preterm pigs, and show how the immature GIT responds to delivery method and different nutritional and therapeutic interventions. The preterm pig may also provide a sensitive model for postnatal adaptation of weak term piglets showing high mortality. Attributes of the preterm pig model include close similarities with preterm infants in body size, organ development, and many clinical features, thereby providing a translational advantage relative to rodent models of GIT immaturity. On the other hand, the need for a sow surgical facility, a piglet intensive care unit, and clinically trained personnel may limit widespread use of preterm pigs. Studies on organ adaptation in preterm pigs help to identify the physiological basis of neonatal survival for hypersensitive newborns and aid in defining the optimal diet and rearing conditions during the critical neonatal period.
Collapse
Affiliation(s)
- P T Sangild
- Department of Nutrition, Exercise, and Sports
| | | | | | | | | | | |
Collapse
|
11
|
Premature delivery reduces intestinal cytoskeleton, metabolism, and stress response proteins in newborn formula-fed pigs. J Pediatr Gastroenterol Nutr 2013; 56:615-22. [PMID: 23364244 DOI: 10.1097/mpg.0b013e318288cf71] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Preterm infants often show intolerance to the first enteral feeds, and the structural and functional basis of this intolerance remains unclear. We hypothesized that preterm and term neonates show similar gut trophic responses to feeding but different expression of intestinal functional proteins, thus helping to explain why preterm neonates are more susceptible to feeding-induced disorders such as necrotizing enterocolitis (NEC). METHODS Incidence of feeding-induced NEC, intestinal mass, and brush border enzyme activities, and the intestinal proteome in preterm cesarean-delivered pigs were compared with the corresponding values in pigs delivered spontaneously at term. RESULTS For both preterm and term pigs, mucosal mass and maltase activity increased (50%-100%), whereas lactase decreased (-50%), relative to values at birth. Only preterm pigs were highly NEC sensitive (30% vs 0% in term pigs, P < 0.05). By gel-based proteomics, 36 identified proteins differed in expression, with most proteins showing downregulation in preterm pigs, including proteins related to intestinal structure and actin filaments, stress response, protein processing, and nutrient metabolism. CONCLUSIONS Despite that enteral feeding induces rapid gut tropic response in both term and preterm neonates, the expression level of cellular proteins related to mucosal integrity, metabolism, and stress response differed markedly (including complement 3, prohibitin, ornithine carbamoyltransferase, and arginosuccinate synthetase). These proteins may play a role in the development of functional gut disorders and NEC in preterm neonates.
Collapse
|
12
|
Abstract
BACKGROUND Changes in the intestinal and colonic proteome in patients with necrotizing enterocolitis (NEC) may help to characterize the disease pathology and identify new biomarkers and treatment targets for NEC. METHODS Using gel-based proteomics, proteins in NEC-affected intestinal and colonic sections were compared with those in adjacent, near-normal tissue sections within the same patients. Western blot and immunohistochemistry were applied to crossvalidate proteomic data and histological location of some selected proteins. RESULTS Thirty proteins were identified with differential expression between necrotic and vital small-intestine sections and 23 proteins were identified for colon sections. Five proteins were similarly affected in the small intestine and colon: histamine receptors (HRs), actins, globins, immunoglobulin, and antitrypsin. Two heat shock proteins (HSPs) were affected in the small intestine. Furthermore, proteins involved in antioxidation, angiogenesis, cytoskeleton formation, and metabolism were affected. Finally, secretory proteins such as antitrypsin, fatty-acid binding protein 5, and haptoglobin differed between NEC-affected and vital tissues. CONCLUSION NEC progression affects different pathways in the small intestine and colon. HSPs may play an important role, especially in the small intestine. The identified secretory proteins should be investigated as possible circulating markers of NEC progression in different gut regions.
Collapse
|
13
|
Antibiotics increase gut metabolism and antioxidant proteins and decrease acute phase response and necrotizing enterocolitis in preterm neonates. PLoS One 2012; 7:e44929. [PMID: 23028687 PMCID: PMC3441690 DOI: 10.1371/journal.pone.0044929] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 08/10/2012] [Indexed: 01/25/2023] Open
Abstract
Background The appropriate use of antibiotics for preterm infants, which are highly susceptible to develop necrotizing enterocolitis (NEC), is not clear. While antibiotic therapy is commonly used in neonates with NEC symptoms and sepsis, it remains unknown how antibiotics may affect the intestine and NEC sensitivity. We hypothesized that broad-spectrum antibiotics, given immediately after preterm birth, would reduce NEC sensitivity and support intestinal protective mechanisms. Methodology/Principal Findings Preterm pigs were treated with antibiotics for 5 d (oral and systemic doses of gentamycin, ampicillin and metrodinazole; AB group) and compared with untreated pigs. Only the untreated pigs showed evidence of NEC lesions and reduced digestive function, as indicated by lowered villus height and activity of brush border enzymes. In addition, 53 intestinal and 22 plasma proteins differed in expression between AB and untreated pigs. AB treatment increased the abundance of intestinal proteins related to carbohydrate and protein metabolism, actin filaments, iron homeostasis and antioxidants. Further, heat shock proteins and the complement system were affected suggesting that all these proteins were involved in the colonization-dependent early onset of NEC. In plasma, acute phase proteins (haptoglobin, complement proteins) decreased, while albumin, cleaved C3, ficolin and transferrin increased. Conclusions/Significance Depressed bacterial colonization following AB treatment increases mucosal integrity and reduces bacteria-associated inflammatory responses in preterm neonates. The plasma proteins C3, ficolin, and transferrin are potential biomarkers of the colonization-dependent NEC progression in preterm neonates.
Collapse
|
14
|
Jiang P, Wan JMF, Sit WH, Lee CL, Schmidt M, Sangild PT. Enteral feeding in utero induces marked intestinal structural and functional proteome changes in pig fetuses. Pediatr Res 2011; 69:123-8. [PMID: 21045752 DOI: 10.1203/pdr.0b013e3182042874] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Intestinal adaptation from parenteral to enteral nutrition is crucial for survival and growth of newborns. Rapid feeding-induced gut maturation occurs immediately after birth in both preterm and term neonates, but it remains unclear whether the responses depend on factors related to birth transition (e.g. bacterial colonization, endocrine, and metabolic changes). We hypothesized that enteral feeding matures the immature intestine, even in fetuses before birth. Hence, control pig fetuses were compared with fetuses fed with milk formula for 24 h in utero. Gel-based proteomics showed that feeding-induced changes in 38 proteins, along with marked increases in intestinal mass and changes in activities of brush border enzymes. Physiological functions of the identified proteins were related to enterocyte apoptosis (e.g. caspase 1) and nutrient metabolism (e.g. citric acid cycle proteins). Many of the differentiated proteins were similar to those identified previously in preterm pigs fed with the same formula after birth, except that effects on proteins related to inflammatory lesions (e.g. heat shock proteins) were absent. Our results show that enteral feeding, independently of the birth transition, induces marked gut maturation and proteome change in the immature intestine. Hence, immediate postnatal feeding-induced gut changes are largely independent of factors related to the birth transition.
Collapse
Affiliation(s)
- Pingping Jiang
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | | | | | | | | | | |
Collapse
|
15
|
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: 5.9] [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
|
16
|
|
17
|
Wang X, Wu W, Lin G, Li D, Wu G, Wang J. Temporal Proteomic Analysis Reveals Continuous Impairment of Intestinal Development in Neonatal Piglets with Intrauterine Growth Restriction. J Proteome Res 2009; 9:924-35. [DOI: 10.1021/pr900747d] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Xiaoqiu Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China 100193, and Department of Animal Science, Texas A&M University, College Station, Texas 77843
| | - Weizong Wu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China 100193, and Department of Animal Science, Texas A&M University, College Station, Texas 77843
| | - Gang Lin
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China 100193, and Department of Animal Science, Texas A&M University, College Station, Texas 77843
| | - Defa Li
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China 100193, and Department of Animal Science, Texas A&M University, College Station, Texas 77843
| | - Guoyao Wu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China 100193, and Department of Animal Science, Texas A&M University, College Station, Texas 77843
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China 100193, and Department of Animal Science, Texas A&M University, College Station, Texas 77843
| |
Collapse
|
18
|
Proteomics at the center of nutrigenomics: Comprehensive molecular understanding of dietary health effects. Nutrition 2009; 25:1085-93. [DOI: 10.1016/j.nut.2009.05.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Accepted: 05/31/2009] [Indexed: 11/18/2022]
|