1
|
Wang Y, Hang C, Hu J, Li C, Zhan C, Pan J, Yuan T. Role of gut-brain axis in neurodevelopmental impairment of necrotizing enterocolitis. Front Neurosci 2023; 17:1059552. [PMID: 36743802 PMCID: PMC9894661 DOI: 10.3389/fnins.2023.1059552] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 01/02/2023] [Indexed: 01/20/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a common gastrointestinal disease of preterm infants with high morbidity and mortality. In survivors of NEC, one of the leading causes of long-term morbidity is the development of severe neurocognitive injury. The exact pathogenesis of neurodevelopmental delay in NEC remains unknown, but microbiota is considered to have dramatic effects on the development and function of the host brain via the gut-brain axis. In this review, we discuss the characteristics of microbiota of NEC, the impaired neurological outcomes, and the role of the complex interplay between the intestinal microbiota and brain to influence neurodevelopment in NEC. The increasing knowledge of microbial-host interactions has the potential to generate novel therapies for manipulating brain development in the future.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neonatology, Children’s Hospital of Zhejiang University, Hangzhou, China
| | - Chengcheng Hang
- Department of Neonatology, Children’s Hospital of Zhejiang University, Hangzhou, China
| | - Jun Hu
- Department of Surgical Intensive Care Unit, Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Chen Li
- Department of Neonatology, Children’s Hospital of Zhejiang University, Hangzhou, China
| | - Canyang Zhan
- Department of Neonatology, Children’s Hospital of Zhejiang University, Hangzhou, China
| | - Jiarong Pan
- Department of Neonatology, Children’s Hospital of Zhejiang University, Hangzhou, China
| | - Tianming Yuan
- Department of Neonatology, Children’s Hospital of Zhejiang University, Hangzhou, China,*Correspondence: Tianming Yuan,
| |
Collapse
|
2
|
Donda KT, Torres BA, Khashu M, Maheshwari A. Single Nucleotide Polymorphisms in Neonatal Necrotizing Enterocolitis. Curr Pediatr Rev 2022; 18:197-209. [PMID: 35040407 DOI: 10.2174/1573396318666220117091621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/10/2021] [Accepted: 11/16/2021] [Indexed: 11/22/2022]
Abstract
The etiopathogenesis of necrotizing enterocolitis (NEC) remains unclear, but increasing information suggests that the risk and severity of NEC may be influenced by single nucleotide polymorphisms in many genes. In this article, we have reviewed gene variations that have either been specifically identified in NEC or have been noted in other inflammatory bowel disorders with similar histopathological abnormalities. We present evidence from our own peer-reviewed laboratory studies and data from an extensive literature search in the databases PubMed, EMBASE, and Scopus. To avoid bias in the identification of existing studies, search keywords were short-listed both from our own studies and from PubMed's Medical Subject Heading (MeSH) thesaurus.
Collapse
Affiliation(s)
- Keyur T Donda
- Department of Pediatrics, University of South Florida Health Morsani College of Medicine, Tampa, FL, USA
| | - Benjamin A Torres
- Department of Pediatrics, University of South Florida Health Morsani College of Medicine, Tampa, FL, USA
| | - Minesh Khashu
- Poole Hospital NHS Foundation Trust and Bournemouth University, Poole, United Kingdom
| | - Akhil Maheshwari
- Department of Pediatrics, Johns Hopkins University, Baltimore, ML, USA
| |
Collapse
|
3
|
Zhang Y, Li K. [Association of interleukin-23 receptor and interleukin-17 single nucleotide polymorphisms with necrotizing enterocolitis in Chinese Han preterm infants]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2020; 22:141-145. [PMID: 32051081 PMCID: PMC7390012 DOI: 10.7499/j.issn.1008-8830.2020.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/16/2020] [Indexed: 06/10/2023]
Abstract
OBJECTIVE To study the association of single nucleotide polymorphisms (SNPs) of interleukin-23 receptor (IL-23R) rs10889677, interleukin-17A (IL-17A) rs227591, and interleukin-17F (IL-17F) rs763780 with necrotizing enterocolitis (NEC) in Chinese Han preterm infants. METHODS A total of 100 Chinese Han preterm infants with NEC who were admitted to the neonatal intensive care unit from January 2017 to January 2019 were prospectively enrolled. Of the 100 preterm infants, 63 had stage II NEC and 37 had stage III NEC. A total of 100 preterm infants, matched for age and sex, were selected as the control group. PCR and Sanger sequencing were used to determine the SNPs of rs10889677, rs2275913, and rs763780. An unconditional logistic regression analysis was used to investigate the association of SNPs with NEC susceptibility and severity. RESULTS The genotype and allele frequencies of rs10889677 and rs2275913 had no influence on the development of NEC (P>0.05). The genotype of rs763780 had no influence on the development of NEC (P>0.05), but the risk of NEC in the infants carrying C allele was 1.652 times that in those carrying T allele (95%CI: 1.052-2.695, P<0.05). The risk of NEC in the infants carrying TC+CC genotype was 1.856 times that in those carrying TT genotype (95%CI: 1.045-3.201, P<0.05). The risk of stage III NEC in the infants carrying TC+CC genotype was 2.965 times that in those carrying TT genotype (95%CI: 1.052-6.330, P<0.05). The risk of stage III NEC in the infants carrying C allele was 2.363 times that in those carrying T allele (95%CI: 1.034-4.093, P<0.05). CONCLUSIONS The SNPs of IL-23R rs10889677 and IL-17A rs2275913 are not associated with the susceptibility to NEC in Chinese Han preterm infants, while TC+CC genotype and C allele of IL-17F rs763780 are associated with the susceptibility to NEC and the severity of NEC.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Neonatology, Women's and Children's Hospital of Qinghai Province, Xining 810000, China.
| | | |
Collapse
|
4
|
Moonen RM, Huizing MJ, González-Luis GE, Cavallaro G, Mosca F, Villamor E. Risk of Necrotizing Enterocolitis Associated With the Single Nucleotide Polymorphisms VEGF C-2578A, IL-18 C-607A, and IL-4 Receptor α -Chain A-1902G: A Validation Study in a Prospective Multicenter Cohort. Front Pediatr 2020; 8:45. [PMID: 32133331 PMCID: PMC7039854 DOI: 10.3389/fped.2020.00045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 01/28/2020] [Indexed: 12/17/2022] Open
Abstract
The etiology of necrotizing enterocolitis (NEC) is multifactorial and an underlying genetic predisposition to NEC is increasingly being recognized. A growing number of studies identified single nucleotide polymorphisms (SNPs) of selected genes with potential biological relevance in the development of NEC. However, few of these genetic studies have been replicated in validation cohorts. We aimed to confirm in a cohort of 358 preterm newborns (gestational age <30 weeks, 26 cases of NEC ≥ Bell stage II) the association with NEC of three candidate SNPs: the vascular endothelium growth factor (VEGF) C-2578A polymorphism (rs699947), the interleukin (IL)-18 C-607A polymorphism (rs1946518), and the IL-4 receptor α-chain (IL-4Rα) A-1902G polymorphism (rs1801275). We observed that allele and genotype frequencies of the three SNPs did not significantly differ between the infants with and without NEC. In contrast, the minor G-allele of the IL-4Rα A-1902G polymorphism was significantly less frequent in the group of 51 infants with the combined outcome NEC or death before 34 weeks postmenstrual age than in the infants without the outcome (0.206 vs. 0.331, P = 0.01). In addition, a significant negative association of the G-allele with the combined outcome NEC or death was found using the dominant (adjusted odds ratio, aOR: 0.44, 95% CI 0.21-0.92), recessive (aOR 0.15, 95% CI 0.03-0.74), and additive (aOR 0.46, 95% CI 0.26-0.80) genetic models. In conclusion our study provides further evidence that a genetic variant of the IL-4Rα gene may contribute to NEC.
Collapse
Affiliation(s)
- Rob M Moonen
- Department of Pediatrics, Zuyderland Medical Center, Heerlen, Netherlands.,Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, Netherlands
| | - Maurice J Huizing
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, Netherlands
| | - Gema E González-Luis
- Department of Pediatrics, Hospital Universitario Materno-Infantil de Canarias, Las Palmas de Gran Canaria, Spain
| | - Giacomo Cavallaro
- Neonatal Intensive Care Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Fabio Mosca
- Neonatal Intensive Care Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, Netherlands
| |
Collapse
|
5
|
Abstract
Necrotizing enterocolitis (NEC) is a devastating disease of prematurity, with no current method for early diagnosis. Diagnosis is particularly challenging, frequently occurring after the disease has progressed to the point of significant and often irreversible intestinal damage. Biomarker research has tremendous potential to advance clinical management of NEC and our understanding of its pathogenesis. This review discusses the need for novel biomarkers in NEC management, evaluates studies investigating such biomarkers, and explains the difficulties associated with translating biomarker discovery into clinical use.
Collapse
Affiliation(s)
- Gregory P Goldstein
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA 94304
| | - Karl G Sylvester
- Department of Surgery, Division of Pediatric Surgery, 300 Pasteur Drive, Alway Building M116, MC 5733, Stanford, CA 94305.
| |
Collapse
|
6
|
Cuna A, George L, Sampath V. Genetic predisposition to necrotizing enterocolitis in premature infants: Current knowledge, challenges, and future directions. Semin Fetal Neonatal Med 2018; 23:387-393. [PMID: 30292709 PMCID: PMC6626706 DOI: 10.1016/j.siny.2018.08.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The role of genetics in the pathogenesis of necrotizing enterocolitis (NEC) was initially informed by epidemiological data indicating differences in prevalence among different ethnic groups as well as concordance in twins. These early observations, together with major advances in genomic research, paved the way for studies that begin to reveal the contribution of genetics to NEC. Using the candidate gene or pathway approach, several potential pathogenic variants for NEC in premature infants have already been identified. More recently, genome-wide association studies and exome-sequencing based studies for NEC have been reported. These advances, however, are tempered by the lack of adequately powered replication cohorts to validate the accuracy of these discoveries. Despite many challenges, genetic research in NEC is expected to increase, providing new insights into its pathogenesis and bringing the promise of personalized care closer to reality. In this review we provide a summary of genetic studies in NEC along with defining the challenges and possible future approaches.
Collapse
Affiliation(s)
| | | | - Venkatesh Sampath
- Division of Neonatology, Department of Pediatrics, Children's Mercy Kansas City, Kansas City, MO, USA.
| |
Collapse
|
7
|
Esdal HCD, Ghbeis MB, Saltzman DA, Hess D, Hume JR, Reed RC, Berry SA, Hoggard E, Hirsch B, Baughn LB, Schimmenti LA. Necrotizing Enterocolitis in Two Siblings and an Unrelated Infant with Overlapping Chromosome 6q25 Deletions. Mol Syndromol 2018; 9:141-148. [PMID: 29928179 DOI: 10.1159/000488817] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2018] [Indexed: 11/19/2022] Open
Abstract
The pathogenesis of necrotizing enterocolitis (NEC) remains poorly understood but is thought to be multifactorial. There are no specific recurring chromosomal abnormalities previously associated with NEC. We report 3 cases of intestinal necrosis associated with large chromosome 6 deletions. The first patient was found to have a 7.9-Mb deletion of chromosome 6 encompassing over 40 genes, arr[GRCh37] 6q25.3q26(155699183_163554531)×1. The second patient had a 19.5-Mb deletion of chromosome 6 generated by an unbalanced translocation with chromosome 18, 46,XY,der(6)t (6;18)(q25.1;p11.23), arr[GRCh37] 6q25.1q27(151639526_ 171115067)×1, 18p11.32p11.23(131700_7694199)×3, which included the whole 7.9-Mb region deleted in the first patient. The third patient was the younger sibling of the second patient with an identical derivative chromosome 6. The shared abnormal chromosome 6 region includes multiple genes of interest, particularly EZR. Mouse models have demonstrated that Ezr is expressed in microvillar epithelium and helps regulate cell-cell adhesion in the gut. We hypothesize that deletion of this shared region of 6q leads to gastrointestinal vulnerability which may predispose patients to intestinal necrosis.
Collapse
Affiliation(s)
- Hannah C D Esdal
- Department of Pediatrics, University of Minnesota, Minneapolis, USA
| | - Muhammad B Ghbeis
- Division of Cardiovascular Critical Care, Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | - Daniel A Saltzman
- Department of Pediatric Surgery, Divisions of, University of Minnesota Masonic Children's Hospital, Minneapolis, USA
| | - Donavon Hess
- Department of Pediatric Surgery, Divisions of, University of Minnesota Masonic Children's Hospital, Minneapolis, USA
| | - Janet R Hume
- Critical Care, University of Minnesota Masonic Children's Hospital, Minneapolis, USA
| | - Robyn C Reed
- Department of Pathology, Children's Hospitals and Clinics of Minnesota, Minneapolis, MN, USA
| | - Susan A Berry
- Genetics and Metabolism, Department of Pediatrics, University of Minnesota Masonic Children's Hospital, Minneapolis, USA
| | - Eric Hoggard
- Division of Pediatric Radiology, Department of Radiology, University of Minnesota Masonic Children's Hospital, Minneapolis, USA
| | - Betsy Hirsch
- Division of Molecular Pathology and Genomics, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, USA
| | - Linda B Baughn
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Lisa A Schimmenti
- Department of Otorhinolaryngology and Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
8
|
Moschopoulos C, Kratimenos P, Koutroulis I, Shah BV, Mowes A, Bhandari V. The Neurodevelopmental Perspective of Surgical Necrotizing Enterocolitis: The Role of the Gut-Brain Axis. Mediators Inflamm 2018; 2018:7456857. [PMID: 29686534 PMCID: PMC5866871 DOI: 10.1155/2018/7456857] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 01/22/2018] [Accepted: 02/05/2018] [Indexed: 02/07/2023] Open
Abstract
This state-of-the-art review article aims to highlight the most recent evidence about the therapeutic options of surgical necrotizing enterocolitis, focusing on the molecular basis of the gut-brain axis in relevance to the neurodevelopmental outcomes of primary peritoneal drainage and primary laparotomy. Current evidence favors primary laparotomy over primary peritoneal drainage as regards neurodevelopment in the surgical treatment of necrotizing enterocolitis. The added exposure to inhalational anesthesia in infants undergoing primary laparotomy is an additional confounding variable but requires further study. The concept of the gut-brain axis suggests that bowel injury initiates systemic inflammation potentially affecting the developing central nervous system. Signals about microbes in the gut are transduced to the brain and the limbic system via the enteric nervous system, autonomic nervous system, and hypothalamic-pituitary axis. Preterm infants with necrotizing enterocolitis have significant differences in the diversity of the microbiome compared with preterm controls. The gut bacterial flora changes remarkably prior to the onset of necrotizing enterocolitis with a predominance of pathogenic organisms. The type of initial surgical approach correlates with the length of functional gut and microbiome equilibrium influencing brain development and function through the gut-brain axis. Existing data favor patients who were treated with primary laparotomy over those who underwent primary peritoneal drainage in terms of neurodevelopmental outcomes. We propose that this is due to the sustained injurious effect of the remaining diseased and necrotic bowel on the developing newborn brain, in patients treated with primary peritoneal drainage, through the gut-brain axis and probably not due to the procedure itself.
Collapse
Affiliation(s)
- Chariton Moschopoulos
- 1Department of Pediatrics, Flushing Hospital Medical Center, SUNY-Stonybrook School of Medicine, Flushing, NY, USA
| | - Panagiotis Kratimenos
- 2Division of Neonatology and Center for Research in Neuroscience, Children's National Medical Center, George Washington University School of Medicine, Washington, DC, USA
| | - Ioannis Koutroulis
- 3Department of Emergency Medicine, Children's National Medical Center, George Washington University School of Medicine, Washington, DC, USA
| | - Bhairav V. Shah
- 4Division of Pediatric Surgery, Palmetto Health Children's Hospital, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Anja Mowes
- 5St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Vineet Bhandari
- 5St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
9
|
Garg BD, Sharma D, Bansal A. Biomarkers of necrotizing enterocolitis: a review of literature. J Matern Fetal Neonatal Med 2017; 31:3051-3064. [PMID: 28756708 DOI: 10.1080/14767058.2017.1361925] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Necrotizing enterocolitis (NEC) is among the most serious gastrointestinal emergency in very low birth weight (VLBW), extremely low birth weight (ELBW) and extremely low gestational age neonates (ELGAN), affecting 7-14% of these neonates. Despite extensive research, the underlying aetiology of NEC still remains blurred. Due to high mortality, morbidity and its delayed presentation, early detection of NEC is considered to be lifesaving. A number of biomarkers have been studied for early detection and prediction of severity of NEC but till date, no ideal marker has been discovered. Molecular techniques like proteomic and metabolomic have recently emerged in the field for the development of biomarkers for early detection and understanding the pathophysiology of NEC. We did literature search for identifying all biomarkers that have been used for the detection of NEC and, in this review article, we discuss these biomarkers along with the available current evidence.
Collapse
Affiliation(s)
- Bhawan Deep Garg
- a Department of Neonatology , Surya Children's Medicare Pvt. Ltd , Mumbai , India
| | - Deepak Sharma
- b Department of Neonatology , National Institute of Medical Sciences , Jaipur , India
| | - Anju Bansal
- c Department of Surgery , Aadrash Hospital , Sri Ganganagar , India
| |
Collapse
|
10
|
Candidate gene analysis in pathogenesis of surgically and non-surgically treated necrotizing enterocolitis in preterm infants. Mol Cell Biochem 2017; 439:53-63. [PMID: 28770467 PMCID: PMC5794808 DOI: 10.1007/s11010-017-3135-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/26/2017] [Indexed: 12/14/2022]
Abstract
Necrotizing enterocolitis (NEC) is one of the most severe and unpredictable complications of prematurity. There are two possible mechanisms involved in the pathogenesis of NEC: individual inflammatory response and impaired blood flow in mesenteric vessels with secondary ischemia of the intestine. The aim of this study was to evaluate the possible relationship between polymorphisms: Il-1β 3953C>T, Il-6 -174G>C and -596G>A, TNFα -308G>A, and 86 bp variable number tandem repeat polymorphism of interleukin-1 receptor antagonist (Il-1RN VNTR 86 bp) and three polymorphisms that may participate in arteries tension regulation and in consequence in intestine blood flow impairment: eNOS (894G>T and -786T>C) and END-1 (5665G>T) and NEC in 100 infants born from singleton pregnancy, before 32 + 0 weeks of gestation, exposed to antenatal steroids therapy, and without congenital abnormalities. In study population, 22 (22%) newborns developed NEC. Surgery-requiring NEC was present in 7 children. Statistical analysis showed 20-fold higher prevalence of NEC in infants with the genotype TT [OR 20 (3.71-208.7); p = 0.0004] of eNOS 894G>T gene polymorphism. There was a higher prevalence of allele C carriers of eNOS 786T>C in patients with surgery-requiring NEC [OR 4.881 (1.33-21.99); p = 0.013]. Our investigation did not confirm any significant prevalence for NEC development in another studied genotypes/alleles. This study confirms the significant role of polymorphisms that play role in intestine blood flow. Identifying gene variants that increase the risk for NEC development may be useful in screening infants with inherent vulnerability and creating strategies for individualized care.
Collapse
|
11
|
Tian J, Liu Y, Jiang Y, Zhou H, Zhu T, Zhao X, Peng L, Yan C. Association of single nucleotide polymorphisms of IL23R and IL17 with necrotizing enterocolitis in premature infants. Mol Cell Biochem 2017; 430:201-209. [PMID: 28224332 DOI: 10.1007/s11010-017-2972-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 02/02/2017] [Indexed: 12/30/2022]
Abstract
Necrotizing enterocolitis (NEC) is a severe gastrointestinal inflammatory disease in neonates, particularly in preterm infants. The interleukin (IL) 23/IL17 axis has been shown to play an important role in the gastrointestinal inflammation. However, the association of gene polymorphisms in the IL23/IL17 axis and the development of NEC remains unknown. In this study, we aimed to explore a possible genetic role of IL23R and IL17 in the development of NEC. We identified single nucleotide polymorphisms (SNPs) in IL23R (rs10889677), IL17A (rs2275913), and IL17F (rs763780) by polymerase chain reaction and Sanger sequencing. A total of 102 NEC patients (stage II, n = 75; and stage III, n = 27) and 120 control subjects were recruited for the study. All of the participants were premature (gestational age < 37 weeks). Our results revealed that the combination of the IL17F rs763780 (TC + CC) genotype and the C allele both significantly increased the risk of NEC [odds ratio (OR) 1.89, 95% confidence interval (CI) 1.04-3.43, P = 0.035; OR 1.82, 95% CI 1.06-3.13, P = 0.028, respectively]. Furthermore, the rs763780 (TC + CC) genotype was associated with increased severity of NEC and the incidence of NEC-related perforation [OR 2.80, 95% CI 1.10-7.12, P = 0.031; OR 3.86, 95% CI 1.10-13.53, P = 0.035, respectively]. However, IL23R rs10889677 and IL17A rs2275913 were not associated with the susceptibility to NEC. In conclusion, our data suggest that a variant of IL17F (rs763780) may contribute to the development of NEC.
Collapse
Affiliation(s)
- Jiayi Tian
- Department of Neonatology, The First Hospital of Jilin University, Changchun, China.,Department of Central Laboratory, The First Hospital of Jilin University, Changchun, China
| | - Yanjun Liu
- Department of Neonatology, The First Hospital of Jilin University, Changchun, China.,Division of Endocrinology, Metabolism, and Molecular Medicine, Charles R. Drew University of Medicine and Sciences, University of California Los Angeles (UCLA) School of Medicine, Los Angeles, CA, USA
| | - Yanfang Jiang
- Department of Central Laboratory, The First Hospital of Jilin University, Changchun, China
| | - Haohan Zhou
- Department of Central Laboratory, The First Hospital of Jilin University, Changchun, China.,Department of Orthopedics Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Tong Zhu
- Department of Neonatology, The First Hospital of Jilin University, Changchun, China
| | - Xiaoqi Zhao
- Department of Neonatology, The Second Hospital of Jilin University, Changchun, China
| | - Liping Peng
- Department of Central Laboratory, The First Hospital of Jilin University, Changchun, China. .,Department of Respirology, The First Hospital of Jilin University, Changchun, China.
| | - Chaoying Yan
- Department of Neonatology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
12
|
Abstract
An underlying genetic predisposition to necrotizing enterocolitis (NEC) is increasingly being recognized. Candidate gene or pathway approaches as well as genome-wide approaches are beginning to identify potential pathogenic variants for NEC in premature infants. However, a majority of these studies have not yielded definitive results because of limited sample size and lack of validation. Despite these challenges, understanding the contribution of genetic variation to NEC is important for providing new insights into the pathogenesis of NEC as well as allowing for targeted care of infants with inherent susceptibility. In this review we provide a summary of published genetic association studies in NEC along with defining the challenges and possible future approaches.
Collapse
Affiliation(s)
- Alain Cuna
- Division of Neonatology, Department of Pediatrics, Children's Mercy Hospital, University of Missouri-Kansas City, 2401 Gillham Rd, Kansas City, MO 64108
| | - Venkatesh Sampath
- Division of Neonatology, Department of Pediatrics, Children's Mercy Hospital, University of Missouri-Kansas City, 2401 Gillham Rd, Kansas City, MO 64108.
| |
Collapse
|
13
|
Hay S, Zupancic JAF, Flannery DD, Kirpalani H, Dukhovny D. Should we believe in transfusion-associated enterocolitis? Applying a GRADE to the literature. Semin Perinatol 2017; 41:80-91. [PMID: 27866662 DOI: 10.1053/j.semperi.2016.09.021] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Numerous observational studies appear to demonstrate an association between packed red blood cell (pRBC) transfusions and necrotizing enterocolitis (NEC). However, the limited numbers of randomized controlled trials (RCTs) do not support a causal relationship between pRBC transfusion and NEC. We sought to determine the quality of the evidence behind transfusion-associated necrotizing enterocolitis (TANEC), and to formulate a GRADE-based recommendation regarding transfusion practices to reduce the risk of TANEC. A systematic search including MEDLINE, Embase, CINAHL, the Cochrane Central Register of Controlled Trials and clinical trials registries was performed for studies assessing the association between transfusion and NEC. Teams of two paired reviewers independently screened studies for eligibility, assessed risk of bias using the GRADE framework, and collected data from each eligible study. We examined studies for two time points following transfusion: within 48h if this was available, and otherwise at any time after transfusion. In total, 23 observational studies and three RCTs met inclusion criteria. The average rating for the quality of evidence of individual studies was between "very low" and "low." On pooling studies for GRADE review, we observed an inconsistency of results. This led to a final overall quality of "very low" for the evidence for an association between transfusions and necrotizing enterocolitis. The pooled outcome of NEC for observational/case control studies was an odds ratio of 1.13 (95% CI: 0.99-1.29) when TANEC was defined as occurring within 48 hours of transfusion. For NEC occurring at any time post-transfusion, the pooled OR was 1.95 (1.60-2.38). Conversely, the pooled outcome of NEC for the RCT data had an odds ratio of 0.6 (0.3, 1.21) with NEC being less frequent in the liberal transfusion group compared to the restrictive transfusion group. The overall quality of the evidence for TANEC is "very low," suggesting very little confidence in the effect estimate. RCT data tended toward apparent protection against NEC. The available evidence is not sufficient to support a practice recommendation around pRBC transfusions in the context of preventing the development of NEC.
Collapse
Affiliation(s)
- Susanne Hay
- Department of Neonatology, Beth Israel Deaconess Medical Center, Boston MA; Department of Pediatrics, Harvard Medical School, Boston, MA; Division of Newborn Medicine, Boston Children's Hospital, Boston, MA
| | - John A F Zupancic
- Department of Neonatology, Beth Israel Deaconess Medical Center, Boston MA; Department of Pediatrics, Harvard Medical School, Boston, MA; Division of Newborn Medicine, Boston Children's Hospital, Boston, MA
| | - Dustin D Flannery
- Division of Neonatology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Haresh Kirpalani
- Division of Neonatology, The Children's Hospital of Philadelphia, Philadelphia, PA; Emeritus Professor, Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada
| | - Dmitry Dukhovny
- Department of Pediatrics, Oregon Health & Science University, Mail Code CDRC-P, 707 SW Gaines St, Portland, OR 97239.
| |
Collapse
|
14
|
|
15
|
Moonen RM, Cavallaro G, Huizing MJ, González-Luis GE, Mosca F, Villamor E. Association between the p.Thr1406Asn polymorphism of the carbamoyl-phosphate synthetase 1 gene and necrotizing enterocolitis: A prospective multicenter study. Sci Rep 2016; 6:36999. [PMID: 27833157 PMCID: PMC5105130 DOI: 10.1038/srep36999] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/20/2016] [Indexed: 12/04/2022] Open
Abstract
The p.Thr1406Asn (rs1047891) polymorphism of the carbamoyl-phosphate synthetase 1 (CPS1) gene has been linked to functional consequences affecting the downstream availability of the nitric oxide precursor L-arginine. L-arginine concentrations are decreased in preterm infants with necrotizing enterocolitis (NEC). In this multicenter prospective study, we investigated the association of the p.Thr1406Asn polymorphism with NEC in 477 preterm infants (36 cases of NEC) from 4 European neonatal intensive care units (Maastricht, Las Palmas de Gran Canaria, Mantova, and Milan). Allele and genotype frequencies of the p.Thr1406Asn polymorphism did not significantly differ between the infants with and without NEC. In contrast, the minor A-allele was significantly less frequent in the group of 64 infants with the combined outcome NEC or death before 34 weeks of corrected gestational age than in the infants without the outcome (0.20 vs. 0.31, P = 0.03). In addition, a significant negative association of the A-allele with the combined outcome NEC or death was found using the dominant (adjusted odds ratio, aOR: 0.54, 95% CI 0.29–0.99) and the additive (aOR 0.58, 95% CI 0.36–0.93) genetic models. In conclusion, our study provides further evidence that a functional variant of the CPS1 gene may contribute to NEC susceptibility.
Collapse
Affiliation(s)
- Rob M Moonen
- Department of Pediatrics, Zuyderland Medical Center Heerlen, 6130 MB, The Netherlands.,Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, 6202 AZ, The Netherlands
| | - Giacomo Cavallaro
- Neonatal Intensive Care Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, 20122, Italy
| | - Maurice J Huizing
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, 6202 AZ, The Netherlands
| | - Gema E González-Luis
- Department of Pediatrics, Hospital Universitario Materno-Infantil de Canarias, Las Palmas de Gran Canaria, 35016, Spain
| | - Fabio Mosca
- Neonatal Intensive Care Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, 20122, Italy
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, 6202 AZ, The Netherlands
| |
Collapse
|
16
|
Niño DF, Sodhi CP, Hackam DJ. Necrotizing enterocolitis: new insights into pathogenesis and mechanisms. Nat Rev Gastroenterol Hepatol 2016; 13:590-600. [PMID: 27534694 PMCID: PMC5124124 DOI: 10.1038/nrgastro.2016.119] [Citation(s) in RCA: 344] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Necrotizing enterocolitis (NEC) is the most frequent and lethal disease of the gastrointestinal tract of preterm infants. At present, NEC is thought to develop in the premature host in the setting of bacterial colonization, often after administration of non-breast milk feeds, and disease onset is thought to be due in part to a baseline increased reactivity of the premature intestinal mucosa to microbial ligands as compared with the full-term intestinal mucosa. The increased reactivity leads to mucosal destruction and impaired mesenteric perfusion and partly reflects an increased expression of the bacterial receptor Toll-like receptor 4 (TLR4) in the premature gut, as well as other factors that predispose the intestine to a hyper-reactive state in response to colonizing microorganisms. The increased expression of TLR4 in the premature gut reflects a surprising role for this molecule in the regulation of normal intestinal development through its effects on the Notch signalling pathway. This Review will examine the current approach to the diagnosis and treatment of NEC, provide an overview of our current knowledge regarding its molecular underpinnings and highlight advances made within the past decade towards the development of specific preventive and treatment strategies for this devastating disease.
Collapse
MESH Headings
- Animals
- Biological Factors/therapeutic use
- Biomarkers/metabolism
- Breast Feeding
- Disease Models, Animal
- Disease Susceptibility
- Enterocolitis, Necrotizing/diagnosis
- Enterocolitis, Necrotizing/etiology
- Enterocolitis, Necrotizing/prevention & control
- Gastrointestinal Microbiome/physiology
- Humans
- Infant, Newborn
- Infant, Premature
- Infant, Premature, Diseases/diagnosis
- Infant, Premature, Diseases/etiology
- Infant, Premature, Diseases/therapy
- Probiotics/therapeutic use
- Treatment Outcome
Collapse
Affiliation(s)
- Diego F Niño
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, Maryland 21287, USA
- The Bloomberg Children's Center, 1800 Orleans Street, Baltimore, Maryland 21287, USA
| | - Chhinder P Sodhi
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, Maryland 21287, USA
- The Bloomberg Children's Center, 1800 Orleans Street, Baltimore, Maryland 21287, USA
| | - David J Hackam
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, Maryland 21287, USA
- The Bloomberg Children's Center, 1800 Orleans Street, Baltimore, Maryland 21287, USA
| |
Collapse
|
17
|
Abstract
Necrotising enterocolitis (NEC) is an uncommon, but devastating intestinal inflammatory disease that predominantly affects preterm infants. NEC is sometimes dubbed the spectre of neonatal intensive care units, as its onset is insidiously non-specific, and once the disease manifests, the damage inflicted on the baby's intestine is already disastrous. Subsequent sepsis and multi-organ failure entail a mortality of up to 65%. Development of effective treatments for NEC has stagnated, largely because of our lack of understanding of NEC pathogenesis. It is clear, however, that NEC is driven by a profoundly dysregulated immune system. NEC is associated with local increases in pro-inflammatory mediators, e.g. Toll-like receptor (TLR) 4, nuclear factor-κB, tumour necrosis factor, platelet-activating factor (PAF), interleukin (IL)-18, interferon-gamma, IL-6, IL-8 and IL-1β. Deficiencies in counter-regulatory mechanisms, including IL-1 receptor antagonist (IL-1Ra), TLR9, PAF-acetylhydrolase, transforming growth factor beta (TGF-β)1&2, IL-10 and regulatory T cells likely facilitate a pro-inflammatory milieu in the NEC-afflicted intestine. There is insufficient evidence to conclude a predominance of an adaptive Th1-, Th2- or Th17-response in the disease. Our understanding of the accompanying regulation of systemic immunity remains poor; however, IL-1Ra, IL-6, IL-8 and TGF-β1 show promise as biomarkers. Here, we chart the emerging immunological landscape that underpins NEC by reviewing the involvement and potential clinical implications of innate and adaptive immune mediators and their regulation in NEC.
Collapse
|
18
|
Nadendla K, Sutton A, Kelly D, Dimmitt R, Wilkinson L, Harmon C, Martin C. Quintuplets: 5 Cases of NEC. Are There Other Risk Factors? Fetal Pediatr Pathol 2016; 35:425-433. [PMID: 27551982 DOI: 10.1080/15513815.2016.1214197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To review a case of quintuplets with all babies developing necrotizing enterocolitis. METHODS A retrospective study of preterm quintuplets all developing necrotizing enterocolitis. Clinical outcomes were reviewed. RESULTS Quintuplets were born at 24 weeks gestation. Each baby developed NEC and was treated. One baby died. Currently the remaining 4 infants are on full enteral nutrition. CONCLUSION Further studies are needed to better understand this emerging population of multiple birth pregnancy and the frequency of NEC development.
Collapse
Affiliation(s)
- Kavita Nadendla
- a Surgery , University of Alabama at Birmingham , Birmingham , Alabama , USA
| | - Amelia Sutton
- b Department of Maternal Fetal Medicine , University of Alabama at Birmingham , Birmingham , Alabama , USA
| | - David Kelly
- c Pathology , Children's Hospital of Alabama , Birmingham , Alabama , USA
| | - Reed Dimmitt
- d Gastroenterology and Surgery , University of Alabama at Birmingham , Birmingham , Alabama , USA
| | - Linda Wilkinson
- a Surgery , University of Alabama at Birmingham , Birmingham , Alabama , USA
| | - Carroll Harmon
- e Surgery , University of Buffalo , Buffalo , New York , USA
| | - Colin Martin
- a Surgery , University of Alabama at Birmingham , Birmingham , Alabama , USA
| |
Collapse
|
19
|
Franklin AL, Said M, Cappiello CD, Gordish-Dressman H, Tatari-Calderone Z, Vukmanovic S, Rais-Bahrami K, Luban NLC, Devaney JM, Sandler AD. Are Immune Modulating Single Nucleotide Polymorphisms Associated with Necrotizing Enterocolitis? Sci Rep 2015; 5:18369. [PMID: 26670709 PMCID: PMC4680983 DOI: 10.1038/srep18369] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 11/16/2015] [Indexed: 01/01/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating gastrointestinal emergency. The purpose of this study is to determine if functional single nucleotide polymorphisms (SNPs) in immune-modulating genes pre-dispose infants to NEC. After Institutional Review Board approval and parental consent, buccal swabs were collected for DNA extraction. TaqMan allelic discrimination assays and BglII endonuclease digestion were used to genotype specific inflammatory cytokines and TRIM21. Statistical analysis was completed using logistic regression. 184 neonates were analyzed in the study. Caucasian neonates with IL-6 (rs1800795) were over 6 times more likely to have NEC (p = 0.013; OR = 6.61, 95% CI 1.48–29.39), and over 7 times more likely to have Stage III disease (p = 0.011; OR = 7.13, (95% CI 1.56–32.52). Neonates with TGFβ-1 (rs2241712) had a decreased incidence of NEC-related perforation (p = 0.044; OR = 0.28, 95% CI: 0.08–0.97) and an increased incidence of mortality (p = 0.049; OR = 2.99, 95% CI: 1.01 – 8.86). TRIM21 (rs660) was associated with NEC-related intestinal perforation (p = 0.038; OR = 4.65, 95% CI 1.09–19.78). In premature Caucasian neonates, the functional SNP IL-6 (rs1800795) is associated with both the development and increased severity of NEC. TRIM21 (rs660) and TGFβ-1 (rs2241712) were associated with NEC- related perforation in all neonates in the cohort. These findings suggest a possible genetic role in the development of NEC.
Collapse
Affiliation(s)
- Ashanti L Franklin
- Division of General and Thoracic Surgery, Children's National Health System, 111 Michigan Ave NW, Washington, DC 20010
| | - Mariam Said
- Division of Neonatology, Children's National Health System, Washington, DC, Department of Pediatrics, The George Washington School of Medicine and Health Sciences, 111 Michigan Ave NW, Washington, DC 20010
| | - Clint D Cappiello
- Division of General and Thoracic Surgery, Children's National Health System, 111 Michigan Ave NW, Washington, DC 20010
| | - Heather Gordish-Dressman
- Children's Research Institute, Children's National Health System, Washington, DC, Department of Pediatrics, The George Washington School of Medicine and Health Sciences, 111 Michigan Ave NW, Washington, DC 20010
| | - Zohreh Tatari-Calderone
- Sheikh Zayed Institute, Children's National Health System, Washington, DC, Department of Pediatrics, The George Washington School of Medicine and Health Sciences, 111 Michigan Ave NW, Washington, DC 20010
| | - Stanislav Vukmanovic
- Sheikh Zayed Institute, Children's National Health System, Washington, DC, Department of Pediatrics, The George Washington School of Medicine and Health Sciences, 111 Michigan Ave NW, Washington, DC 20010
| | - Khodayar Rais-Bahrami
- Division of Neonatology, Children's National Health System, Washington, DC, Department of Pediatrics, The George Washington School of Medicine and Health Sciences, 111 Michigan Ave NW, Washington, DC 20010
| | - Naomi L C Luban
- Department Laboratory Medicine, Children's National Health System Washington, DC, Department of Pediatrics, The George Washington School of Medicine and Health Sciences, 111 Michigan Ave NW, Washington, DC 20010
| | - Joseph M Devaney
- Department of Genetic Medicine, Children's National Health System, 111 Michigan Ave NW, Washington, DC 20010
| | - Anthony D Sandler
- Division of General and Thoracic Surgery, Children's National Health System, Washington, DC, Department of Pediatrics, The George Washington School of Medicine and Health Sciences, 111 Michigan Ave NW, Washington, DC 20010
| |
Collapse
|
20
|
Markel TA, Engelstad H, Poindexter BB. Predicting disease severity of necrotizing enterocolitis: how to identify infants for future novel therapies. J Clin Neonatol 2014; 3:1-9. [PMID: 24741531 PMCID: PMC3982330 DOI: 10.4103/2249-4847.128717] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Necrotizing enterocolitis (NEC) remains a very devastating problem within the very low birth weight neonatal population. Several experimental therapies are being tested in animal models and soon may be ready for human trials. Despite this progress, we currently have no way to identify infants who would be optimal targets for therapy. Specifically, we are unable to predict which infants will progress to the more severe Bell's stage of disease that may necessitate surgery. Ideally, an algorithm could be constructed that would encompass multiple neonatal and maternal risk factors as well as potential biologic markers of disease so that these infants could be identified in a more timely fashion. This review summarizes the known risk factors and biomarkers of disease in hopes of stimulating clinical research to identify such an “early warning” NEC algorithm.
Collapse
Affiliation(s)
- Troy A Markel
- Department of Surgery, Section of Pediatric Surgery, Riley Hospital for Children at Indiana University Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Holly Engelstad
- Department of Pediatrics, Section of Neonatology, Riley Hospital for Children at Indiana University Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Brenda B Poindexter
- Department of Pediatrics, Section of Neonatology, Riley Hospital for Children at Indiana University Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
21
|
Sylvester KG, Ling XB, Liu GYG, Kastenberg ZJ, Ji J, Hu Z, Wu S, Peng S, Abdullah F, Brandt ML, Ehrenkranz RA, Harris MC, Lee TC, Simpson BJ, Bowers C, Moss RL. Urine protein biomarkers for the diagnosis and prognosis of necrotizing enterocolitis in infants. J Pediatr 2014; 164:607-12.e1-7. [PMID: 24433829 PMCID: PMC4161235 DOI: 10.1016/j.jpeds.2013.10.091] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 08/21/2013] [Accepted: 10/14/2013] [Indexed: 10/25/2022]
Abstract
OBJECTIVES To test the hypothesis that an exploratory proteomics analysis of urine proteins with subsequent development of validated urine biomarker panels would produce molecular classifiers for both the diagnosis and prognosis of infants with necrotizing enterocolitis (NEC). STUDY DESIGN Urine samples were collected from 119 premature infants (85 NEC, 17 sepsis, 17 control) at the time of initial clinical concern for disease. The urine from 59 infants was used for candidate biomarker discovery by liquid chromatography/mass spectrometry. The remaining 60 samples were subject to enzyme-linked immunosorbent assay for quantitative biomarker validation. RESULTS A panel of 7 biomarkers (alpha-2-macroglobulin-like protein 1, cluster of differentiation protein 14, cystatin 3, fibrinogen alpha chain, pigment epithelium-derived factor, retinol binding protein 4, and vasolin) was identified by liquid chromatography/mass spectrometry and subsequently validated by enzyme-linked immunosorbent assay. These proteins were consistently found to be either up- or down-regulated depending on the presence, absence, or severity of disease. Biomarker panel validation resulted in a receiver-operator characteristic area under the curve of 98.2% for NEC vs sepsis and an area under the curve of 98.4% for medical NEC vs surgical NEC. CONCLUSIONS We identified 7 urine proteins capable of providing highly accurate diagnostic and prognostic information for infants with suspected NEC. This work represents a novel approach to improving the efficiency with which we diagnose early NEC and identify those at risk for developing severe, or surgical, disease.
Collapse
Affiliation(s)
- Karl G. Sylvester
- Division of Pediatric Surgery, Lucile Packard Children’s Hospital, Palo Alto, CA,Department of Surgery, Stanford University School of Medicine, Baltimore, MD
| | - Xuefeng B. Ling
- Department of Surgery, Stanford University School of Medicine, Baltimore, MD
| | - Gigi Yuen-Gee Liu
- Department of Surgery, Stanford University School of Medicine, Baltimore, MD,Pediatric Surgery, Department of Surgery, Johns Hopkins University, Baltimore, MD
| | - Zachary J. Kastenberg
- Division of Pediatric Surgery, Lucile Packard Children’s Hospital, Palo Alto, CA,Department of Surgery, Stanford University School of Medicine, Baltimore, MD
| | - Jun Ji
- Department of Surgery, Stanford University School of Medicine, Baltimore, MD
| | - Zhongkai Hu
- Department of Surgery, Stanford University School of Medicine, Baltimore, MD
| | - Shuaibin Wu
- Department of Surgery, Stanford University School of Medicine, Baltimore, MD
| | - Sihua Peng
- Department of Surgery, Stanford University School of Medicine, Baltimore, MD
| | - Fizan Abdullah
- Pediatric Surgery, Department of Surgery, Johns Hopkins University, Baltimore, MD
| | - Mary L. Brandt
- Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, TX
| | - Richard A. Ehrenkranz
- Department of Pediatrics, Division of Neonatology, Yale University School of Medicine, New Haven, CT
| | - Mary Catherine Harris
- Department of Pediatrics, Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Timothy C. Lee
- Pediatric Surgery, Department of Surgery, Texas Children’s Hospital, Houston, TX
| | - B. Joyce Simpson
- Department of Pediatrics, Division of Neonatology, Yale University School of Medicine, New Haven, CT
| | - Corinna Bowers
- Pediatric Surgery, Department of Surgery, Nationwide Children’s Hospital, Columbus, OH
| | - R. Lawrence Moss
- Pediatric Surgery, Department of Surgery, Nationwide Children’s Hospital, Columbus, OH
| |
Collapse
|
22
|
|
23
|
Abstract
Different categories of biomarkers of necrotising enterocolitis (NEC), including (i) non-specific mediators of the inflammatory cascade, e.g. acute phase reactants, chemokines, cytokines, and cell surface antigens, (ii) enhanced non-specific biomarkers, and (iii) specific gut-associated proteins, have distinctive biochemical characteristics and properties. The appropriateness of using these mediators in specific clinical situations, and the pros and cons of their applications as indicators or predictors of intestinal injury and NEC are highlighted. Many potentially new biomarkers such as micro-RNA, volatile organic compounds and gut microbiomes are currently under investigation. A stringent protocol for biomarker discovery is revealed so that investigators can consider this methodology as a reference for future discovery of organ-specific and/or disease-specific biomarkers for preterm infants.
Collapse
Affiliation(s)
- Pak Cheung Ng
- Department of Paediatrics, 6th Floor, Clinical Sciences Building, Prince of Wales Hospital, Shatin, N.T., Hong Kong.
| |
Collapse
|
24
|
Abstract
Necrotizing enterocolitis (NEC) has largely been present in neonatal intensive care units for the past 60 years. NEC prevalence has corresponded with the continued development and sophistication of neonatal intensive care. Despite major efforts towards its eradication, NEC has persisted and appears to be increasing in some centers. The pathophysiology of this disease remains poorly understood. Several issues have hampered our quest to develop a better understanding of this disease. These include the fact that what we have historically termed 'NEC' appears to be several different diseases. Animal models that are commonly used to study NEC pathophysiology and treatment do not directly reflect the most common form of the disease seen in human infants. The pathophysiology appears to be multifactorial, reflecting several different pathways to intestinal necrosis with different inciting factors. Spontaneous intestinal perforations, ischemic bowel disease secondary to cardiac anomalies as well as other entities that are clearly different from the most common form of NEC seen in preterm infants have been put into the same database. Here I describe some of the different forms of what has been called NEC and make some comments on its pathophysiology, where available studies suggest involvement of genetic factors, intestinal immaturity, hemodynamic instability, inflammation and a dysbiotic microbial ecology. Currently utilized approaches for the diagnosis of NEC are presented and innovative technologies for the development of diagnostic and predictive biomarkers are described. Predictions for future strategies are also discussed.
Collapse
Affiliation(s)
- Josef Neu
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Fla., USA
| |
Collapse
|
25
|
Prencipe G, Auriti C, Inglese R, Gallusi G, Dotta A, De Benedetti F. The macrophage migration inhibitory factor -173G/C polymorphism is not significantly associated with necrotizing enterocolitis in preterm infants. J Pediatr Surg 2013; 48:1499-502. [PMID: 23895962 DOI: 10.1016/j.jpedsurg.2013.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 12/28/2012] [Accepted: 01/04/2013] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND PURPOSE Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality among premature infants. Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine that has been implicated in the pathophysiology of inflammatory bowel diseases. The MIF promoter contains a functionally relevant single nucleotide polymorphism (SNP) G→C at position -173, with the MIF -173*C allele being associated with higher MIF expression in vitro and with higher MIF levels in vivo. The aim of this study was to investigate whether the G/C polymorphism at -173 of the MIF promoter is associated with the development of NEC. METHODS In this retrospective cohort study, 107 preterm infants (GA ≤ 32 weeks), of whom 41 had NEC (NEC Stage I n = 20, Stage II n = 3, Stage III n = 18) and 66 were not affected, were genotyped for the MIF -173 SNP. MIF genotyping was carried out by PCR and DHPLC. RESULTS We did not find significant differences in the prevalence of the -173G/C polymorphism and in the distribution of the -173 MIF genotype in infants with NEC compared to controls. Moreover, we did not observe an association between the polymorphism and mortality. CONCLUSIONS The polymorphism -173G/C of the MIF promoter does not appear to be of major importance in the pathophysiology of NEC in preterm infants.
Collapse
Affiliation(s)
- Giusi Prencipe
- Laboratory of Rheumatology, Bambino Gesù Children's Hospital, Roma, Italy
| | | | | | | | | | | |
Collapse
|
26
|
Ng PC, Chan KYY, Poon TCW. Biomarkers for prediction and diagnosis of necrotizing enterocolitis. Clin Perinatol 2013; 40:149-59. [PMID: 23415270 DOI: 10.1016/j.clp.2012.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This article summarizes the commonly used biomarkers currently available for diagnosis of necrotizing enterocolitis. The most exciting advances in diagnostic tests were the use of new nucleic acid sequencing techniques (eg, next-generation sequencing) and molecular screening methods (eg, proteomics and microarray analysis) for the discovery of novel biomarkers. The new technology platform coupled with stringent protocols of biomarker discovery and validation would enable neonatologists to study biologic systems at a level never before possible and discover unique biomarkers for specific organ injury and/or disease entity.
Collapse
Affiliation(s)
- Pak C Ng
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin NT, Hong Kong SAR, The People's Republic of China.
| | | | | |
Collapse
|
27
|
Abstract
OBJECTIVE The objective of this study was to compare demographic and clinical events in three groups of preterm neonates: those with necrotizing enterocolitis totalis (NEC-T), those with NEC non-totalis (NEC non-T) and in preterm patients without NEC. STUDY DESIGN This retrospective case-control study was conducted at Yale New Haven Children's Hospital using patient data from January 1991 to December 2007. Study patients were less than 36 weeks of gestational age (GA) at birth, without gastrointestinal (GI) malformations. Cases (NEC-T) were diagnosed at operation or at autopsy with observation of >80% necrosis of the GI tract. Two control groups were assigned: Group 1 or NEC non-T and Group II or Non-NEC. Two to four controls per case were matched to cases by GA at birth±2 weeks. Demographic and clinical data for the day of diagnosis and retrospectively up to 7 days preceding diagnosis were recorded for those with NEC-T and NEC. Group II controls were matched for date of birth and day of life, in addition to GA at birth. RESULT A total of 14 075 patients were admitted to the Newborn Special Care Unit during the study interval. Overall 328 patients (2.3%) developed NEC≥Bell's Stage II; 39 patients met inclusion criteria for NEC-T case status; 148 NEC non-T and 110 non-NEC controls were assigned. In the comparison of NEC T and NEC non-T neonates, use of breast milk was associated with decreased risk of NEC-T, adjusted odds ratio (OR)=0.26, 95% confidence interval (CI) of OR=0.08-0.085, P=0.03. When NEC T and non-NEC patients were compared, having reached full-enteral feeds before the date of diagnosis of the matched case (adjusted OR=28.5, 95% CI of OR=2.7-299, P=0.005) and use of breast milk (adjusted OR=0.09, 95% CI of OR=0.02-0.56, P=0.01) were significantly different between the two groups. CONCLUSION Breast milk usage was significantly associated with decreased occurrence of NEC-T in our comparison of NEC-T, NEC non-T and non-NEC patients. Although there were some differences, the majority of demographic and clinical variables assessed were not shown to be significantly different between cases and controls. This highlights the need for more biological data in assessing risk of developing NEC-T.
Collapse
|
28
|
The NFKB1 (g.-24519delATTG) variant is associated with necrotizing enterocolitis (NEC) in premature infants. J Surg Res 2011; 169:e51-7. [PMID: 21529841 DOI: 10.1016/j.jss.2011.03.017] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 02/11/2011] [Accepted: 03/04/2011] [Indexed: 01/04/2023]
Abstract
OBJECTIVE While it is known that gene-environment interactions contribute to necrotizing enterocolitis (NEC) pathogenesis, characterization of genetic risk-factors that can predict NEC in preterm infants remains nascent. We hypothesized that altered intestinal immune responses arising from sequence variation in the toll-like receptor (TLR) pathway genes contribute to NEC susceptibility. MATERIALS AND METHODS Very low birth weight (VLBW) infants were recruited prospectively in a multi-center, cohort study involving collection of blood samples along with collation of clinical information. DNA obtained from blood samples was used to genotype nine single nucleotide polymorphisms (SNPs) in eight TLR pathway genes by single-base extension. Prevalence of the variant allele was compared between cases and controls using Fisher's exact test. RESULTS In our cohort of 271 infants, 15 infants (5.6%) developed NEC, and five died from it. Infants with NEC were less mature (P < 0.001), and were more likely to be African-American (P = 0.007). SNPs in the TLR2, TLR4, TLR5, TLR9, IRAK1, and TIRAP genes were not associated with NEC. The NFKB1 (g.-24519delATTG) variant was present in all infants with NEC but only in 65% of infants without NEC (P = 0.003), while the NFKBIA (g.-1004A>G) variant was present in 13.3% of infants with NEC but in 49% of infants without NEC (P = 0.007). After correcting for multiple comparisons, the NFKB1 and NFKBIA variants remained associated with NEC (P < 0.05). CONCLUSIONS These data suggest that TLR genetic variants can alter susceptibility to NEC in VLBW infants and support the hypothesis that genetically programmed differences in the innate immune response contribute to NEC pathogenesis.
Collapse
|
29
|
Abstract
During developmental age, differences in pharmacodynamic reactions to several drugs may reflect polymorphisms of genes encoding drug-transporting proteins, receptors, drug targets, and gene products, whose disturbed activity sometimes plays an important role in certain diseases. Administration of drugs with a narrow therapeutic index may quite easily be associated with changes in pharmacokinetics and development of adverse drug reactions, which occasionally may cause fatalities. In such cases, polypragmasy and resulting drug interactions may enhance effects of changes in drug-metabolizing enzymes' activities. Phenotyping and genotyping of patients slowly are finding their place in some therapeutic regimens used in clinical gastroenterology and hepatology. At present, some assays to measure, for example, thiopurine S-methyltransferase activity are already commercially available. Polymorphisms of CYP450 enzymes, interleukins, and altered gene expression play an important role in some patients' various gastrointestinal tract and liver diseases. Herbal drugs also affect proinflammatory and antiinflammatory cytokine and nitric oxide balance in the body. Therapeutic use of recombined proteins, such as infliximab, natalizumab, onercept, humanized antibody to integrin α-4 β-7, or IFN-β in some large-bowel diseases increased therapeutic efficacy. IFN-α used in the patients with chronic hepatitis C improved cellular immunity in these subjects and exerted antiviral activity. Practical application of progress in pharmacogenetics, pharmacokinetics, pharmacodynamics, and use of bioproducts in novel therapeutic regimens has opened therapeutic frontiers and increased clinical safety.
Collapse
|
30
|
Abstract
Neonatal septic shock is a devastating condition associated with high morbidity and mortality. Definitions for the sepsis continuum and treatment algorithms specific for premature neonates are needed to improve studies of septic shock and assess benefit from clinical interventions. Unique features of the immature immune system and pathophysiologic responses to sepsis, particularly those of extremely preterm infants, necessitate that clinical trials consider them as a separate group. Keen clinical suspicion and knowledge of risk factors will help to identify those neonates at greatest risk for development of septic shock. Genomic and proteomic approaches, particularly those that use very small sample volumes, will increase our understanding of the pathophysiology and direct the development of novel agents for prevention and treatment of severe sepsis and shock in the neonate. Although at present antimicrobial therapy and supportive care remain the foundation of treatment, in the future immunomodulatory agents are likely to improve outcomes for this vulnerable population.
Collapse
|
31
|
Christensen RD, Lambert DK, Henry E, Wiedmeier SE, Snow GL, Baer VL, Gerday E, Ilstrup S, Pysher TJ. Is “transfusion-associated necrotizing enterocolitis” an authentic pathogenic entity? Transfusion 2009; 50:1106-12. [DOI: 10.1111/j.1537-2995.2009.02542.x] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
32
|
Young C, Sharma R, Handfield M, Mai V, Neu J. Biomarkers for infants at risk for necrotizing enterocolitis: clues to prevention? Pediatr Res 2009; 65:91R-97R. [PMID: 19190533 PMCID: PMC2929681 DOI: 10.1203/pdr.0b013e31819dba7d] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Necrotizing enterocolitis (NEC) is the most common severe gastrointestinal emergency that affects premature newborns. This disease often has a rapid onset with few, if any, antecedent signs that can be used to reliably predict its occurrence. Its rapid onset and progression to death, as well as its severe morbidity when the infant survives, begs for early diagnostic tools that may be used in determining those infants who would be at greatest risk for development of the disease and for whom early preventative measures could be targeted. Although studies have suggested efficacy of several techniques such as breath hydrogen, inflammatory mediators in blood, urine or stool, and genetic markers, these all have drawbacks limiting their use. The application of newly developed "omic" approaches may provide biomarkers for early diagnosis and targeted prevention of this disease.
Collapse
Affiliation(s)
- Christopher Young
- Department of Pediatrics, University of Florida, Gainesville, Florida 32610
| | - Renu Sharma
- Department of Pediatrics, University of Florida, Jacksonville, Florida 32209
| | - Martin Handfield
- Department of Oral Biology and Center for Molecular Microbiology, University of Florida, Gainesville, Florida 32610
| | - Volker Mai
- Department of Microbiology, University of Florida, Gainesville, Florida 32610
| | - Josef Neu
- Department of Pediatrics, University of Florida, Gainesville, Florida 32610
| |
Collapse
|
33
|
Lin PW, Nasr TR, Stoll BJ. Necrotizing enterocolitis: recent scientific advances in pathophysiology and prevention. Semin Perinatol 2008; 32:70-82. [PMID: 18346530 DOI: 10.1053/j.semperi.2008.01.004] [Citation(s) in RCA: 202] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality among infants in the neonatal intensive care unit. Here we review the epidemiology and pathophysiology of NEC, with an emphasis on the latest research findings and potential areas for future research. NEC continues to be one of the most devastating and unpredictable diseases affecting premature infants. Despite decades of research, the pathogenesis of this disease remains unclear, and prevention and treatment strategies are limited. Hopefully, future studies aimed at understanding premature intestinal defenses, commensal or probiotic bacterial influences, and possible genetic predisposition will lead to the improvement of prevention and treatment strategies.
Collapse
Affiliation(s)
- Patricia W Lin
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.
| | | | | |
Collapse
|
34
|
Abstract
In necrotizing enterocolitis (NEC) the small (most often distal) and/or large bowel becomes injured, develops intramural air, and may progress to frank necrosis with perforation. Even with early, aggressive treatment, the progression of necrosis, which is highly characteristic of NEC, can lead to sepsis and death. This article reviews the current scientific knowledge related to the etiology and pathogenesis of NEC and discusses some possible preventive measures.
Collapse
|
35
|
Abstract
Necrotizing enterocolitis (NEC) is a devastating intra-abdominal emergency in the newborn period. The disease involves bowel wall inflammation, ischemic necrosis, eventual perforation, and the need for urgent surgical intervention. Unrecognized or left untreated, the neonate can decompensate quickly, often progressing to shock, multisystem organ failure, and eventual death. During the past several years, a number of basic science and clinical trials have been established in an attempt to understand the pathophysiology of NEC. As many researchers feel that NEC develops as an uncontrolled inflammatory response that leads to intestinal ischemia, a large number of studies have been focused on the inflammatory cascade and the role that cytokines play within that cascade. Although a large amount of data has been generated from these studies, the events leading to the ischemic injury of the intestine are still not fully understood. This article will therefore focus on the key cytokines involved with NEC, in an attempt to present the current literature and studies that support their involvement.
Collapse
Affiliation(s)
- Troy A Markel
- Department of Surgery, Indiana University School of Medicine, the Indiana Center for Vascular Biology and Medicine, Indianapolis, IN, USA
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
Bronchopulmonary dysplasia (BPD) is a common perinatal complication of very low birth weight preterm infants with a significant risk of long-term disability and morbidity. While clinical conditions such as prematurity and mechanical ventilation are its major risk factors, studies suggest that there is an individual susceptibility to BPD. This comprehensive review summarizes data collected about the implication of genetic polymorphisms in BPD and in its risk factors. Some studies have directly related the risk of BPD to genotype. Indeed, carrier states of genetic variants of cytokines (IFNgamma T+874A), adhesion molecules (L-selectin-Pro213Ser), elements of renin-angiotensin system (ACE-I/D), antioxidant enzymes (GST-P1 Val105Ile), and surfactant proteins (SPA1, SPB intron 4) has been identified as risk factors to BPD. Other studies investigated the role of genotype in BPD risk factors. Premature birth has been linked to carrier states of genetic variants with an impact on immune status (such as IL-6 G(-174)C, MBL2 54G/A, VEGF G+405C, HSP72 A+1267G genes) and matrix metalloproteases. Fetal inflammatory response syndrome, a major determinant of BPD is also affected by genotype (including LTalpha A+250G). Disturbed intrauterine lung development and vascularization may also contribute to BPD; these processes may be impaired in the presence of some rare genetic mutations. Furthermore, there is also a genetic component in the susceptibility to other perinatal adaptational disturbances such as respiratory distress syndrome that are associated with an increased need for mechanical ventilation, and, hence, with lung damage. The genetic variants presented in this article may help to identify infants at risk for BPD.
Collapse
Affiliation(s)
- Géza Bokodi
- Ist Department of Pediatrics, Semmelweis University, Budapest, Hungary.
| | | | | | | | | |
Collapse
|
37
|
Abstract
The main aim of identifying gene-environment interactions is to provide insight into mechanisms of disease development and to identify patients with an inherent vulnerability to certain conditions. This in turn may allow patients to be targeted with individualised treatment based on the knowledge of their inborn susceptibility to specific conditions. This review describes the possible effects of common genetic variation on outcome in various conditions affecting the neonate. It focuses predominantly on studies of positive association rather than non-association to illustrate this potential influence and to highlight the potential for further study and intervention. The shortcomings of published association studies and the place of such studies in future research are also discussed.
Collapse
MESH Headings
- Chronic Disease
- DNA/genetics
- Developmental Disabilities/genetics
- Enterocolitis, Necrotizing/genetics
- Gene Expression/genetics
- Genetic Predisposition to Disease/genetics
- Humans
- Infant, Newborn
- Infant, Newborn, Diseases/genetics
- Infant, Newborn, Diseases/immunology
- Infant, Premature/physiology
- Infant, Small for Gestational Age/physiology
- Lung Diseases/genetics
- Polymorphism, Genetic/genetics
- Renal Insufficiency/genetics
- Research Design
- Respiratory Distress Syndrome, Newborn/genetics
- Sepsis/genetics
Collapse
Affiliation(s)
- David Harding
- University of Bristol, D Level, St Michael's Hospital, Bristol BS2 8EG, UK.
| |
Collapse
|
38
|
Moonen RMJ, Paulussen ADC, Souren NYP, Kessels AGH, Rubio-Gozalbo ME, Villamor E. Carbamoyl phosphate synthetase polymorphisms as a risk factor for necrotizing enterocolitis. Pediatr Res 2007; 62:188-90. [PMID: 17597649 DOI: 10.1203/pdr.0b013e3180a0324e] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
A C-to-A nucleotide transversion (T1405N) in the gene that encodes carbamoyl-phosphate synthetase 1 (CPS1) has been correlated with low plasma concentrations of L-arginine in neonates. As plasma L-arginine concentrations are decreased in premature infants with necrotizing enterocolitis (NEC), we hypothesized that the CPS1 T1405N polymorphism would correlate with the presence of NEC. We analyzed the CPS1 genotypes for the T1405N polymorphism in 17 preterm infants (<or=30 wk and <1500 g) with established NEC, 34 preterm infants without NEC, and 25 healthy term infants. Distribution of genotypes did not differ between the NEC population (CC:AC:AA = 70.6%:23.5%:5.9%) and the preterm control group (CC:AC:AA = 41.2%:35.3%:23.5%; p = 0.110) or the term group (CC:AC:AA = 44%:48%:8%; p = 0.228). The C allele frequency was 82.4% in NEC and 58.8% in preterm control infants (p = 0.018) and analysis for linear trend demonstrated that incidence of NEC increased with the number of C alleles (p = 0.037). The CC genotype was associated with an increased risk of NEC in the preterm infants [odds ratio (OR) = 3.43, 95% confidence interval (CI): 1.01-11.49, p = 0.048), when compared with the grouped together AA/AC genotypes. These data suggest that the CPS1 T1405N polymorphism may be associated with the risk of NEC in preterm infants.
Collapse
Affiliation(s)
- Rob M J Moonen
- Department of Pediatrics, University Hospital of Maastricht, 6202 AZ Maastricht, The Netherlands
| | | | | | | | | | | |
Collapse
|
39
|
Abstract
Necrotising enterocolitis is one of the most common gastrointestinal emergencies in newborn infants. Here we review the epidemiology, clinical presentation, and pathophysiology of the disease, as well as strategies for diagnosis, management, and prevention. Necrotising enterocolitis is one of the most devastating and unpredictable diseases affecting premature infants. Despite decades of research, its pathogenesis remains unclear; diagnosis can be difficult; and treatment is challenging. We will need to improve our understanding of intestinal defences in premature infants, dietary and bacterial factors, and genetic effects that could predispose infants to necrotising enterocolitis before we can develop new strategies for prevention and treatment.
Collapse
Affiliation(s)
- Patricia W Lin
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | | |
Collapse
|
40
|
Martin CR, Walker WA. Intestinal immune defences and the inflammatory response in necrotising enterocolitis. Semin Fetal Neonatal Med 2006; 11:369-77. [PMID: 16690363 DOI: 10.1016/j.siny.2006.03.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Necrotising enterocolitis is a devastating neonatal gastrointestinal emergency predominantly affecting low birth weight, premature infants and is accompanied by significant mortality and morbid sequelae. The pathophysiology remains obscure and the management of infants with necrotising enterocolitis has not changed since the recognition of this disease. Necrotising enterocolitis is most likely the clinical culmination of multiple different risk factors interacting with each other to produce bowel injury through a final, common inflammatory pathway. Here, we review intestinal immunity and the specific inflammatory mediators involved in this disease process.
Collapse
MESH Headings
- Enterocolitis, Necrotizing/immunology
- Enterocolitis, Necrotizing/mortality
- Enterocolitis, Necrotizing/pathology
- Humans
- Infant, Newborn
- Infant, Premature
- Infant, Premature, Diseases/immunology
- Infant, Premature, Diseases/mortality
- Infant, Premature, Diseases/pathology
- Intestines/immunology
- Prevalence
- Risk Factors
Collapse
Affiliation(s)
- Camilia R Martin
- Harvard Medical School, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Rose-318, Boston, MA 02215, USA.
| | | |
Collapse
|
41
|
Bokodi G, Derzbach L, Vásárhelyi B. Re: Deletion allele of angiotensin-converting enzyme. J Pediatr 2006; 149:579; author reply 579-80. [PMID: 17011342 DOI: 10.1016/j.jpeds.2006.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2006] [Accepted: 05/03/2006] [Indexed: 11/25/2022]
|
42
|
Bhandari V, Bizzarro MJ, Shetty A, Zhong X, Page GP, Zhang H, Ment LR, Gruen JR. Familial and genetic susceptibility to major neonatal morbidities in preterm twins. Pediatrics 2006; 117:1901-6. [PMID: 16740829 DOI: 10.1542/peds.2005-1414] [Citation(s) in RCA: 269] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Intraventricular hemorrhage, necrotizing enterocolitis, and bronchopulmonary dysplasia remain significant causes of morbidity and mortality in preterm newborns. OBJECTIVES Our goal was to assess the familial and genetic susceptibility to intraventricular hemorrhage, necrotizing enterocolitis, and bronchopulmonary dysplasia. METHODS Mixed-effects logistic-regression and latent variable probit model analysis were used to assess the contribution of several covariates in a multicenter retrospective study of 450 twin pairs born at < or =32 weeks of gestation. To determine the genetic contribution, concordance rates in a subset of 252 monozygotic and dizygotic twin pairs were compared. RESULTS The study population had a mean gestational age of 29 weeks and birth weight of 1286 g. After controlling for effects of covariates, the twin data showed that 41.3%, 51.9%, and 65.2%, respectively, of the variances in liability for intraventricular hemorrhage, necrotizing enterocolitis, and bronchopulmonary dysplasia could be accounted for by genetic and shared environmental factors. Among the 63 monozygotic twin pairs, the observed concordance for bronchopulmonary dysplasia was significantly higher than the expected concordance; 12 of 18 monozygotic twin pairs with > or =1 affected member had both members affected versus 3.69 expected. After controlling for covariates, genetic factors accounted for 53% of the variance in liability for bronchopulmonary dysplasia. CONCLUSIONS Twin analyses show that intraventricular hemorrhage, necrotizing enterocolitis, and bronchopulmonary dysplasia are familial in origin. These data demonstrate, for the first time, the significant genetic susceptibility for bronchopulmonary dysplasia in preterm infants.
Collapse
Affiliation(s)
- Vineet Bhandari
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520-8064, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Neu J, Chen M, Beierle E. Intestinal innate immunity: how does it relate to the pathogenesis of necrotizing enterocolitis. Semin Pediatr Surg 2005; 14:137-44. [PMID: 16084400 DOI: 10.1053/j.sempedsurg.2005.05.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The pathogenesis of necrotizing enterocolitis (NEC) is poorly understood, but appears to be multifactorial and highly associated with immaturity of the gastrointestinal tract, colonization of the intestinal microbiota, and immature innate immune system. The goal of this review is to provide an overview of some of these risk factors and how they might lead to the genesis of NEC. A better understanding of these factors should help us prevent and treat this devastating disease.
Collapse
Affiliation(s)
- Josef Neu
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida 32608, USA
| | | | | |
Collapse
|
44
|
Upperman JS, Potoka D, Grishin A, Hackam D, Zamora R, Ford HR. Mechanisms of nitric oxide-mediated intestinal barrier failure in necrotizing enterocolitis. Semin Pediatr Surg 2005; 14:159-66. [PMID: 16084403 DOI: 10.1053/j.sempedsurg.2005.05.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Necrotizing enterocolitis (NEC) is the leading intestinal emergency in premature infants. The underlying etiology of NEC remains elusive, but hypoxic conditions and early enteral feeding are consistently implicated as the main risk factors in the pathogenesis of NEC. We postulate that nitric oxide (NO) plays a key role as a molecular signaling "hub" in the generation of gut barrier failure in NEC. Clinical studies suggest that inflammatory cytokines and excessive NO production may contribute to the pathogenesis of NEC. One of the major challenges in defining the critical signaling pathways that lead to the development of NEC is the lack of specific biochemical markers that consistently delineate the early stages of NEC. Intestinal pathology and molecular markers derived from late-stage NEC represent end-stage findings and thus provide little insight into the early events that led to intestinal inflammation. Such markers may not represent viable therapeutic targets for the treatment or prevention of NEC. Therefore, novel strategies are needed to identify the patients at risk for NEC and define the clinically relevant molecules that characterize the early stages of NEC. This review will examine the mechanisms of NO-mediated gut barrier failure and propose novel genetic-based approaches for elucidating the critical molecular pathways in NEC.
Collapse
Affiliation(s)
- Jeffrey S Upperman
- Department of Surgery, University of Pittsburgh and Children's Hospital of Pittsburgh, Pittsburgh, Pennsylania 15213, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Vásárhelyi B, Tóth-Heyn P, Treszl A, Tulassay T. Genetic polymorphisms and risk for acute renal failure in preterm neonates. Pediatr Nephrol 2005; 20:132-5. [PMID: 15627170 DOI: 10.1007/s00467-004-1711-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2004] [Revised: 09/16/2004] [Accepted: 09/17/2004] [Indexed: 11/24/2022]
Abstract
Acute renal failure (ARF) affects about 10% of severely ill neonates. Recent studies have shown that genetic polymorphisms of proteins that play a role in neonatal physiology may contribute to individual susceptibility to both ARF and its risk factors. Our review summarizes the data collected to date. Studies have shown that the risk of preterm neonates for ARF is directly associated with a combination of high tumor necrosis factor-alpha producer and low interleukin-6 producer genotypes, as well as with low heat shock protein 72 producer genotype. Premature birth is itself the most important risk factor for a number of complications, including ARF, and recent studies have also shown an association between several maternal and fetal cytokine genetic polymorphisms and increased inflammatory response in preterm neonates. These polymorphisms could also be associated with increased risk for disorders such as sepsis and necrotizing enterocolitis, which lead to renal hypoperfusion and ARF. Genetic polymorphisms of the renin-angiotensin-aldosterone system have not been shown to directly influence risk for ARF. They may, however, be associated with patent ductus arteriosus, poor postnatal adaptation, and heart failure, which are all prevalent risk factors for ARF.
Collapse
Affiliation(s)
- Barna Vásárhelyi
- Research Group of Pediatrics and Nephrology, Hungarian Academy of Sciences, Budapest, Hungary
| | | | | | | |
Collapse
|